US20110301087A1 - Crf1 receptor compounds - Google Patents

Crf1 receptor compounds Download PDF

Info

Publication number
US20110301087A1
US20110301087A1 US13/127,602 US200913127602A US2011301087A1 US 20110301087 A1 US20110301087 A1 US 20110301087A1 US 200913127602 A US200913127602 A US 200913127602A US 2011301087 A1 US2011301087 A1 US 2011301087A1
Authority
US
United States
Prior art keywords
residue
absent
compound
seq
alanine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/127,602
Inventor
Edward McBride
Athan Kuliopulos
Thomas J. McMurry
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
ANCHOR THERAPEUTICS Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US13/127,602 priority Critical patent/US20110301087A1/en
Assigned to ANCHOR THERAPEUTICS, INC. reassignment ANCHOR THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MCBRIDE, EDWARD, MCMURRY, THOMAS J., KULIOPULOS, ATHAN
Publication of US20110301087A1 publication Critical patent/US20110301087A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/57509Corticotropin releasing factor [CRF] (Urotensin)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/542Carboxylic acids, e.g. a fatty acid or an amino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/06Antiabortive agents; Labour repressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • G protein coupled receptors constitute one of the largest families of genes in the human genome. GPCRs are integral membrane signaling proteins. Hydrophobicity mapping of the amino acid sequences of G-protein coupled receptors has led to a model of the typical G-protein-coupled receptor as containing seven hydrophobic membrane-spanning regions with the amino terminal on the extracellular side of the membrane and the carboxyl terminal on the intracellular side of the membrane.
  • GPCRs mediate the transmission of intracellular signals (“signal transduction”) by activating guanine nucleotide-binding proteins (G proteins) to which the receptor is coupled. GPCRs are activated by a wide range of endogenous stimuli, including peptides, amino acids. hormones, light, and metal ions. The following reviews are incorporated by reference: Hill, British J. Pharm 147: s27 (2006); Palczeski, Ann Rev Biochemistry 75: 743-767 (2006); Dorsham & Gutkind, Nature Reviews 7: 79-94 (2007); Kobilka & Schertler, Trends Pharmacol Sci. 2: 79-83 (2008).
  • GPCRs are important targets for drug discovery as they are involved in a wide range of cellular signaling pathways and are implicated in many pathological conditions (e.g., cardiovascular and mental disorders, cancer, AIDS). In fact, GPCRs are targeted by 40-50% of approved drugs, illustrating the critical importance of this class of pharmaceutical targets. Interestingly, this number represents only about 30 GPCRs, a small fraction of the total number of GPCRs thought to be relevant to human disease. Over 1000 GPCRs are known in the human genome, and GPCRs remain challenging targets from a research and development perspective in part because these amembrane bound receptors with complex pharmacology.
  • GPCR modulators e.g., agonists, partial agonists, inverse agonists and antagonists
  • allosteric modulators of GPCRs e.g., negative and positive allosteric modulators, allosteric agonists, and ago-allosteric modulators.
  • the invention relates generally to compounds which are allosteric modulators (e.g., negative and positive allosteric modulators, allosteric agonists, and ago-allosteric modulators) of the G protein coupled receptor for corticotrophin releasing hormone (or factor) receptor 1, also known as the CRF1, CRHR1, CRFR1, CRHR, CRF-R.
  • the CRF1 receptor compounds are derived from the intracellular loops and domains of CRF1 receptor.
  • the invention also relates to the use of these CRF1 receptor compounds and pharmaceutical compositions comprising the CRF1 receptor compounds in the treatment of diseases and conditions associated with CRF1 receptor modulation, such as inflammatory bowel disease (peripherally acting), irritable bowel syndrome (IBS), stress response (colonic motor activity), anxiety, sleep disorder, addictive behavior, acute and chronic neurodegeneration, preterm labor and pain.
  • diseases and conditions associated with CRF1 receptor modulation such as inflammatory bowel disease (peripherally acting), irritable bowel syndrome (IBS), stress response (colonic motor activity), anxiety, sleep disorder, addictive behavior, acute and chronic neurodegeneration, preterm labor and pain.
  • the invention also relates to pharmaceutical compositions comprising one or more compounds of the invention and a carrier, and the use of the disclosed compounds and compositions in methods of treating diseases and conditions responsive to modulation of the CRF1 receptor.
  • GPCRs G Protein Coupled Receptors
  • G protein coupled receptors constitute one of the largest superfamilies of genes in the human genome; these transmembrane proteins enable the cell the respond to its environment by sensing extracellular stimuli and initiating intracellular signal transduction cascades. GPCRs mediate signal transduction through the binding and activation of guanine nucleotide-binding proteins (G proteins) to which the receptor is coupled. Wide arrays of ligands bind to these receptors, which in turn orchestrate signaling networks integral to many cellular functions. Diverse GPCR ligands include small proteins, peptides, amino acids, biogenic amines, lipids, ions, odorants and even photons of light. The following reviews are incorporated by reference: Hill, British J. Pharm 147: s27 (2006); Dorsam & Gutkind, Nature Reviews 7: 79-94 (2007).
  • GPCR signaling pathways are integral components of many pathological conditions (e.g., cardiovascular and mental disorders, cancer, AIDS).
  • GPCRs are targeted by 40-50% of approved drugs illustrating the critical importance of this class of pharmaceutical targets.
  • this number represents only about 30 GPCRs, a small fraction of the total number of GPCRs thought to be relevant to human disease.
  • GPCRs are membrane bound receptors that exhibit complex pharmacological properties and remain challenging targets from a research and development perspective. Given their importance in human health combined with their prevalence (over 1000 known GPCRs in the human genome) GPCRs represent an important target receptor class for drug discovery and design.
  • GPCRs are integral membrane proteins that mediate diverse signaling cascades through an evolutionarily conserved structural motif. All GPCRs are thought to consist of seven hydrophobic transmembrane spanning ⁇ -helices with the amino terminus on the extracellular side of the membrane and the carboxyl terminus on the intracellular side of the membrane. The transmembrane helices are linked together sequentially by extracellular (e1, e2, e3) and intracellular (cytoplasmic) loops (i1, i2, i3).
  • the intracellular loops or domains are intimately involved in the coupling and turnover of G proteins and include: i1, which connects TM1-TM2; i2, connecting TM3-TM4; i3, connecting TM5-TM6; and a portion of the C-terminal cytoplasmic tail (domain 4). Due in part to the topological homology of the 7TM domains and the recent high resolution crystal structures of several GPCRs (Palczewski et al., Science 289, 739-45 (2000), Rasmussen, S. G. et al., Nature 450, 383-7 (2007)) skilled modelers are now able to predict the general boundaries of GPCR loop domains through the alignment of several related receptors.
  • GPCR mediated signal transduction is initiated by the binding of a ligand to its cognate receptor.
  • GPCR ligand binding is believed to take place in a hydrophilic pocket generated by a cluster of helices near the extracellular domain.
  • other ligands such as large peptides, are thought to bind to the extracellular region of protein and hydrophobic ligands are postulated to intercalate into a receptor binding pocket through the membrane between gaps in the helices. The process of ligand binding induces conformational changes within the receptor.
  • this process is catalytic and results in signal amplification in that activation of one receptor may elicit the activation and turnover of numerous G proteins, which in turn may regulate multiple second messenger systems.
  • Signaling diversity is further achieved through the existence of numerous G protein types as well as differing isoforms of alpha, beta and gamma subunits.
  • GPCRs interact with G proteins to regulate the synthesis or inhibition of intracellular second messengers such as cyclic AMP, inositol phosphates, diacylglycerol and calcium ions, thereby triggering a cascade of intracellular events that eventually leads to a biological response.
  • GPCR signaling may be modulated and attenuated through cellular machinery as well as pharmacological intervention. Signal transduction may be ‘switched off’ with relatively fast kinetics (seconds to minutes) by a process called rapid desensitization. For GPCRs, this is caused by a functional uncoupling of receptors from heterotrimeric G proteins, without a detectable change in the total number of receptors present in cells or tissues. This process involves the phosphorylation of the receptor C terminus, which enables the protein arrestin to bind to the receptor and occlude further G protein coupling. Once bound by arrestin the receptor may be internalized into the cell and either recycled back to the cell surface or degraded.
  • the alpha subunit of the G protein possesses intrisic GTPase activity, which attenuates signaling and promotes re-association with the beta/gamma subunits and a return to the basal state.
  • GPCR signaling may also be modulated pharmacologically. Agonist drugs act directly to activate the receptors, whereas antagonist drugs act indirectly to block receptor signaling by preventing agonist activity through their associating with the receptor.
  • GPCR binding and signaling can also be modified through allosteric modulation, that is by ligands that bind not at the orthosteric binding site but through binding at an allosteric site elsewhere in the receptors.
  • Allosteric modulators can include both positive and negative modulators of orthosteric ligand mediated activity, allosteric agonists (that act in the absence of the orthosteric ligand), and ago-allosteric modulators (ligands that have agonist activity on their own but that can also modulate the activity of the orthosteric ligand).
  • GPCR families include Class A Rhodopsin like, Class B Secretin like, Class C Metabotropic glutamate/pheromone, Class D Fungal pheromone, Class E cAMP receptors (Dictyostelium), the Frizzled/Smoothened family, and various orphan GPCRs.
  • putative families include Ocular albinism proteins, Insect odorant receptors, Plant Mlo receptors, Nematode chemoreceptors, Vomeronasal receptors (VIR & V3R) and taste receptors.
  • CRF1 is a class B GPCR, also called family B or secretin-like.
  • class B receptors are activated by peptide ligands typically 30 to 40 amino acids in length. Activation of these receptors results in activation of adenylyl cyclase and signal transduction through increase in cAMP as a primary signaling pathway.
  • Class B receptors have a large N-terminal extracellular domain with 4 very highly conserved cysteine residues. This domain is important for the binding of endogenous peptide ligands and resulting receptor activation. While these receptors signal primarily through Gs activation of adenylyl cyclase, they also couple to Gq, resulting in calcium release and may also couple to Gi/GO, which modulate adenylyl cyclase activity.
  • CRF1 corticotrophin releasing hormone
  • urocortin I urocortin I
  • II urocortin II
  • III urocortin I
  • CRF1 activation mediates colonic motility, while activation of the related receptor CRF2 delays gastric emptying.
  • At least 8 isoforms of CRF1 have been identified, but not all isoforms appear to be functional.
  • CRF1-deficient mice possess an impaired stress response and reduced anxiety.
  • P is a peptide comprising at least three contiguous amino-acid residues (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18 or 19) of an intracellular i1, i2 or i3 loop or intracellular i4 domain of the CRF1 receptor.
  • the N-terminal nitrogen of the N-terminal amino acid residue of P to which the linking moiety C(O) is bonded can be one of the at least three contiguous amino acid residues or it can be an amino acid residue distinct from the at least three contiguous amino acid residues.
  • Intracellular i1 loop refers to the loop which connects TM1 to TM2 and the corresponding transmembrane junctional residues.
  • Intracellular i2 loop refers to the loop which connects TM3 to TM4 and the corresponding transmembrane junctional residues.
  • Intracellular i3 loop refers to the loop which connects TM5 to TM6 and the corresponding transmembrane junctional residues.
  • Intracellular i4 domain refers to the C-terminal cytoplasmic tail and the transmembrane junctional residue.
  • P comprises at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, at least fifteen, at least sixteen, at least seventeen, at least eighteen or at least nineteen contiguous amino acid residues of the intracellular i1, i2 or i3 loop or intracellular i4 domain of the CRF1 receptor
  • the at least three contiguous amino acids of P are derived from the intracellular i1, i2 or i3 loop or intracellular i4 domain of the CRF1 receptor, wherein the amino acid sequence of each loop and the i4 domain is as described in Table 1.
  • the intracellular loop for the i1 loop, i2 loop, i3 loop and i4 domain can also include amino acids from transmembrane junctional residues.
  • the i1 loop can include SEQ ID NO: 1 where one or more residues from the transmembrane junctional residues are included on either the C-terminus, the N-terminus or both.
  • SEQ ID NO: 1 can include either an Threonine residue, Leucine residue or both at the C-terminus.
  • P comprises at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, at least fifteen, at least sixteen, at least seventeen, at least eighteen or at least nineteen contiguous amino acid residues of the i1 intracellular loop of the CRF1 receptor.
  • P is derived from the i1 loop and is represented by the following structural formula or pharmaceutically acceptable salts thereof
  • At least four of X 7 , X 8 , X 9 , X 10 , X 11 , X 12 , X 13 and X 14 are present.
  • X 7 is an arginine residue
  • X 8 is a serine residue
  • X 9 is an isoleucine residue
  • X 10 is an arginine residue.
  • X 11 is a cysteine or serine residue
  • X 12 is a leucine residue
  • X 13 is an arginine residue
  • X 14 is an asparagine residue.
  • X 11 is a cysteine residue.
  • X 11 is a serine residue.
  • X 14 is an asparagine residue
  • X 15 is an isoleucine residue
  • X 16 is an isoleucine residue
  • X 17 is a histidine residue.
  • P is selected from the group consisting of SEQ ID NOS: 1-23 as listed in Table 2 below:
  • the at least three contiguous amino acids of P are derived from the i2 intracellular loop of the CRF1 receptor.
  • P derived from the i2 loop and is represented by the following structural formula or a pharmaceutically acceptable salt thereof
  • At least three of Y 7 , Y 8 , Y 9 , Y 10 , Y 11 , Y 12 , Y 13 , Y 14 , Y 15 , Y 16 , Y 17 , Y 18 and Y 19 are present.
  • Y 9 is serine, threonine or glycine residue
  • Y 10 is a threonine, serine, tyrosine, glycine or alanine residue
  • Y 11 is an aspartic acid, alanine, serine or glycine residue.
  • Y 12 is an arginine or alanine residue
  • Y 13 is a leucine or an alanine residue
  • Y 14 is an arginine or alanine residue.
  • Y 9 is serine, threonine or glycine residue
  • Y 10 is a threonine, serine, tyrosine, glycine or alanine residue
  • Y 11 is an aspartic acid, alanine, serine or glycine residue
  • Y 12 is an arginine or alanine residue.
  • Y 11 is an aspartic acid, alanine, serine or glycine residue
  • Y 12 is an arginine or alanine residue
  • Y 13 is a leucine or an alanine residue
  • Y 14 is an arginine or alanine residue.
  • Y 9 is serine, threonine or glycine residue
  • Y 10 is a threonine, serine, tyrosine, glycine or alanine residue
  • Y 11 is an aspartic acid, alanine, serine or glycine residue
  • Y 12 is an arginine or alanine residue
  • Y 13 is a leucine or an alanine residue.
  • Y 10 is a threonine, serine, tyrosine, glycine or alanine residue
  • Y 11 is an aspartic acid, alanine, serine or glycine residue
  • Y 12 is an arginine or alanine residue
  • Y 13 is a leucine or an alanine residue
  • Y 14 is an arginine or alanine residue.
  • Y 9 is serine, threonine or glycine residue
  • Y 10 is a threonine, serine, tyrosine, glycine or alanine residue
  • Y 11 is an aspartic acid, alanine, serine or glycine residue
  • Y 12 is an arginine or alanine residue
  • Y 13 is a leucine or an alanine residue
  • Y 14 is an arginine or alanine residue.
  • Y 9 is serine residue
  • Y 10 is a threonine residue
  • Y 11 is an aspartic acid residue
  • Y 12 is an arginine residue
  • Y 13 is a leucine residue
  • Y 14 is an arginine residue.
  • Y 7 is a threonine or serine residue
  • Y 8 is a tyrosine or glycine residue
  • Y 9 is a serine, threonine or glycine residue
  • Y 10 is a threonine, serine, tyrosine, glycine or alanine residue.
  • Y 7 is a threonine residue
  • Y 8 is a tyrosine residue
  • Y 9 is a serine residue
  • Y 10 is a threonine residue.
  • Y 11 is an aspartic acid, alanine, serine or glycine residue. Specifically, Y 11 is an aspartic acid residue.
  • Y 12 is an arginine or alanine residue
  • Y 13 is or a leucine or an alanine residue
  • Y 14 is an arginine or alanine residue
  • Y 15 is a lysine or an alanine residue.
  • one of Y 12 , Y 13 , Y 14 , and Y 15 is an alanine residue, provided that the other three of Y 12 , Y 13 , Y 14 , and Y 15 are not an alanine residue.
  • Y 12 is an arginine residue
  • Y 13 is a leucine residue
  • Y 14 is an arginine residue
  • Y 15 is a lysine residue.
  • Y 16 is a tryptophan, methionine or an alanine residue. Specifically, Y 16 is a tryptophan residue. Alternatively, Y 16 is a methionine residue.
  • Y 15 is a lysine or an alanine residue
  • Y 16 is a tryptophan, methionine or an alanine residue
  • Y 17 is a methionine, norleucine, phenylalanine or an alanine residue
  • Y 18 is a phenylalanine, isoleucine or an alanine residue.
  • one of Y 15 , Y 16 , Y 17 , and Y 18 is an alanine residue, provided that the other three of Y 15 , Y 16 , Y 17 , and Y 18 are not an alanine residue.
  • Y 15 is a lysine residue
  • Y 16 is a tryptophan or methionine residue
  • Y 17 is a methionine, norleucine, or phenylalanine residue
  • Y 18 is a phenylalanine or isoleucine residue.
  • Y 15 is a lysine residue
  • Y 16 is a tryptophan residue
  • Y 17 is a methionine residue
  • Y 18 is a phenylalanine residue.
  • Y 19 is an isoleucine or alanine residue. Specifically, Y 19 is an isoleucine residue.
  • P is selected from the group consisting of SEQ ID NOS: 24-70 as listed in Table 3 below:
  • P comprises at least three contiguous amino (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or 19) of the i3 intracellular loop of the CRF1 receptor.
  • P is derived from the i3 loop and is represented by the following structural formula or a pharmaceutically acceptable salt thereof:
  • Z 1 is absent or a phenylalanine residue
  • Z 2 is absent or an asparagine residue
  • Z 3 is absent or an isoleucine residue
  • Z 4 is absent or a valine residue
  • Z 5 is absent or an arginine residue
  • Z 6 is absent or an isoleucine residue
  • Z 7 is absent or a leucine residue
  • Z 8 is absent or a methionine residue
  • Z 9 is absent or a threonine residue
  • Z 10 is absent or a lysine residue
  • Z 11 is absent or a leucine residue
  • Z 12 is absent or an arginine residue
  • Z 13 is absent or an alanine residue
  • Z 14 is absent or a serine residue
  • Z 15 is absent or a threonine residue
  • Z 16 is absent or a threonine residue
  • Z 17 is absent or a serine residue
  • Z 18 is absent or a glutamic acid residue
  • Z 19 is absent or a threonine residue
  • Z 20 is absent or an isoleucine residue
  • Z 21 is absent or a glutamine residue
  • Z 22 is absent or a tyrosine residue
  • Z 23 is absent or an arginine residue
  • Z 24 is absent or a lysine residue
  • Z 25 is absent or an alanine residue
  • Z 26 is absent or a valine residue
  • Z 27 is absent or a lysine residue
  • Z 28 is absent or an alanine or serine residue
  • Z 29 is absent or a threonine residue
  • Z 30 is absent or a leucine residue
  • R 1 is OR 2 or N(R 2 ) 2 ;
  • each R 2 is independently hydrogen or (C 1 -C 10 )alkyl
  • Z 1 -Z 30 it is bonded to L as written. For example, Z 1 is bonded to L. If Z 1 is absent, then Z 2 is bonded to L.
  • At least three of Z 5 , Z 6 , Z 7 , Z 8 , and Z 9 are present.
  • Z 5 is an arginine residue
  • Z 6 is an isoleucine residue
  • Z 7 is a leucine residue.
  • Z 7 is a leucine residue
  • Z 8 is a methionine residue
  • Z 9 is a threonine residue.
  • Z 5 is an arginine residue
  • Z 6 is an isoleucine residue
  • Z 7 is a leucine residue
  • Z 8 is a methionine residue.
  • Z 6 is an isoleucine residue
  • Z 7 is a leucine residue
  • Z 8 is absent or a methionine residue
  • Z 9 is a threonine residue.
  • Z 5 is an arginine residue
  • Z 6 is an isoleucine residue
  • Z 7 is a leucine residue
  • Z 8 is a methionine residue
  • Z 9 is a threonine residue.
  • At least three of Z 10 , Z 11 , Z 12 , and Z 13 are present.
  • Z 10 is a lysine residue
  • Z 11 is a leucine residue
  • Z 12 is an arginine residue.
  • Z 11 is a leucine residue
  • Z 12 is an arginine residue
  • Z 13 is an alanine residue.
  • Z 10 is a lysine residue
  • Z 11 is a leucine residue
  • Z 12 is an arginine residue
  • Z 13 is an alanine residue.
  • At least three of Z 14 , Z 15 , Z 16 , and Z 17 are present.
  • Z 14 is a serine residue
  • Z 15 is a threonine residue
  • Z 16 is a threonine residue.
  • Z 15 is a threonine residue
  • Z 16 is a threonine residue
  • Z 17 is a serine residue
  • Z 14 is a serine residue
  • Z 15 is a threonine residue
  • Z 16 is a threonine residue
  • Z 17 is a serine residue
  • At least three of Z 19 , Z 20 , Z 21 , Z 22 , and Z 23 are present.
  • Z 19 is a threonine residue
  • Z 20 is an isoleucine residue
  • Z 21 is a glutamine residue.
  • Z 21 is a glutamine residue
  • Z 22 is a tyrosine residue
  • Z 23 is an arginine residue.
  • Z 19 is a threonine residue
  • Z 20 is an isoleucine residue
  • Z 21 is a glutamine residue
  • Z 22 is a tyrosine residue.
  • Z 20 is an isoleucine residue
  • Z 21 is a glutamine residue
  • Z 22 is a tyrosine residue
  • Z 23 is an arginine residue.
  • Z 19 is a threonine residue
  • Z 20 is an isoleucine residue
  • Z 21 is a glutamine residue
  • Z 22 is a tyrosine residue
  • Z 23 is an arginine residue.
  • At least three of Z 24 , Z 25 , Z 26 , Z 27 , and Z 28 are present.
  • Z 24 is a lysine residue
  • Z 25 is an alanine residue
  • Z 26 is a valine residue.
  • Z 26 is a valine residue
  • Z 27 is a lysine residue
  • Z 28 is an alanine or serine residue.
  • Z 24 is a lysine residue
  • Z 25 is an alanine residue
  • Z 26 is a valine residue
  • Z 27 is a lysine residue.
  • Z 25 is an alanine residue
  • Z 26 is a valine residue
  • Z 27 is a lysine residue
  • Z 28 is an alanine or serine residue.
  • Z 24 is a lysine residue
  • Z 25 is an alanine residue
  • Z 26 is a valine residue
  • Z 27 is a lysine residue
  • Z 28 is an alanine or serine residue. Specifically, Z 28 is an alanine residue.
  • P is selected from the group consisting of SEQ ID NOS: 71-98 as listed in Table 4 below:
  • P comprises at least three contiguous amino (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or 19) of the i4 intracellular domain of the CRF1 receptor.
  • P derived from the i4 domain and is represented by the following structural formula or a pharmaceutically acceptable salt thereof
  • M 1 is absent or a glutamic acid residue
  • M 2 is absent or a valine residue
  • M 3 is absent or an arginine residue
  • M 4 is absent or a serine residue
  • M 5 is absent or an alanine residue
  • M 6 is absent or an isoleucine residue
  • M 7 is absent or an arginine residue
  • M 8 is absent or a lysine residue
  • M 9 is absent or an arginine residue
  • M 10 is absent or a tryptophan residue
  • M 11 is absent or a histidine residue
  • M 12 is absent or an arginine residue
  • M 13 is absent or a tryptophan residue
  • M 14 is absent or a glutamine residue
  • M 15 is absent or an aspartic acid residue
  • M 16 is absent or a lysine residue
  • M 17 is absent or a histidine or glycine residue
  • M 18 is absent or a serine residue
  • M 19 is absent or an isoleucine residue
  • M 20 is absent or an arginine residue
  • M 21 is absent or an alanine residue
  • M 22 is absent or an arginine residue
  • M 23 is absent or a valine residue
  • M 24 is absent or an alanine residue
  • M 25 is absent or an arginine residue
  • M 26 is absent or an alanine residue
  • M 27 is absent or a methionine residue
  • M 28 is absent or a serine residue
  • M 29 is absent or an isoleucine residue
  • M 30 is absent or a proline residue
  • M 31 is absent or a threonine residue
  • M 32 is absent or a serine residue
  • M 33 is absent or a proline residue
  • M 34 is absent or a threonine residue
  • M 35 is absent or an arginine residue
  • M 36 is absent or a valine residue
  • M 37 is absent or a serine residue
  • M 38 is absent or a phenylalanine residue
  • M 39 is absent or a histidine residue
  • M 40 is absent or a serine residue
  • M 41 is absent or an isoleucine residue
  • M 42 is absent or a lysine residue
  • M 43 is absent or a glutamine residue
  • M 44 is absent or a serine residue
  • M 45 is absent or a threonine residue
  • M 46 is absent or an alanine residue
  • M 47 is absent or a valine residue; provided that at least five of M 1 -M 47 are present;
  • R 1 is OR 2 or N(R 2 ) 2 ;
  • each R 2 is independently hydrogen or (C 1 -C 10 )alkyl
  • M 1 -M 47 it is bonded to L as written. For example, M 1 is bonded to L. If M 1 is absent, then M 2 is bonded to L.
  • At least three of M 2 -M 7 are present.
  • M 2 is a valine residue
  • M 3 is an arginine residue
  • M 4 is a serine residue.
  • M 5 is an alanine residue
  • M 6 is an isoleucine residue
  • M 7 is an arginine residue.
  • M 2 is a valine residue
  • M 3 is an arginine residue
  • M 4 is a serine residue
  • M 5 is an alanine residue.
  • M 4 is a serine residue
  • M 5 is an alanine residue
  • M 6 is an isoleucine residue
  • M 7 is an arginine residue.
  • M 2 is a valine residue
  • M 3 is an arginine residue
  • M 4 is a serine residue
  • M 5 is an alanine residue
  • M 6 is an isoleucine residue.
  • M 3 is an arginine residue
  • M 4 is a serine residue
  • M 5 is an alanine residue
  • M 6 is an isoleucine residue
  • M 7 is an arginine residue.
  • M 2 is a valine residue
  • M 3 is an arginine residue
  • M 4 is a serine residue
  • M 5 is an alanine residue
  • M 6 is an isoleucine residue
  • M 7 is an arginine residue.
  • At least three of M 9 -M 13 are present.
  • M 9 is an arginine residue
  • M 10 is a tryptophan residue
  • M 11 is a histidine residue
  • M 12 is an arginine residue.
  • M 10 is a tryptophan residue
  • M 11 is a histidine residue
  • M 12 is an arginine residue
  • M 13 is a tryptophan residue.
  • M 9 is an arginine residue
  • M 10 is a tryptophan residue
  • M 11 is a histidine residue
  • M 12 is an arginine residue
  • M 13 is a tryptophan residue.
  • At least three of M 18 -M 22 are present.
  • M 18 is a serine residue
  • M 19 is an isoleucine residue
  • M 20 is an arginine residue
  • M 21 is an alanine residue.
  • M 19 is an isoleucine residue
  • M 20 is an arginine residue
  • M 21 is an alanine residue
  • M 22 is an arginine residue.
  • M 18 is a serine residue
  • M 19 is an isoleucine residue
  • M 20 is an arginine residue
  • M 21 is an alanine residue
  • M 22 is an arginine residue.
  • At least three of M 23 -M 27 are present.
  • M 23 is a valine residue
  • M 24 is an alanine residue
  • M 25 is an arginine residue
  • M 26 is an alanine residue.
  • M 24 is an alanine residue
  • M 25 is an arginine residue
  • M 26 is an alanine residue
  • M 27 is a methionine residue.
  • M 23 is a valine residue
  • M 24 is an alanine residue
  • M 25 is an arginine residue
  • M 26 is an alanine residue
  • M 27 is a methionine residue.
  • At least three of M 25 -M 29 are present.
  • M 25 is an arginine residue
  • M 26 is an alanine residue
  • M 27 is a methionine residue
  • M 28 is a serine residue.
  • M 26 is an alanine residue
  • M 27 is a methionine residue
  • M 28 is a serine residue
  • M 29 is an isoleucine residue.
  • M 25 is an arginine residue
  • M 26 is an alanine residue
  • M 27 is a methionine residue
  • M 28 is a serine residue
  • M 29 is an isoleucine residue.
  • At least three of M 32 -M 36 are present.
  • M 32 is a serine residue
  • M 33 is a proline residue
  • M 34 is a threonine residue
  • M 35 is an arginine residue.
  • M 33 is a proline residue
  • M 34 is a threonine residue
  • M 35 is an arginine residue
  • M 36 is a valine residue.
  • M 32 is a serine residue
  • M 33 is a proline residue
  • M 34 is a threonine residue
  • M 35 is an arginine residue
  • M 36 is a valine residue.
  • P is selected from the group consisting of SEQ ID NOS: 99-106 as listed in Table 5 below:
  • sequences presented in Tables 2-5 can be optionally functionalized at the C-terminus.
  • Functionalized at the C-terminus means that the acid moiety present at the C-terminus is replaced by some other functional group.
  • Suitable functional groups include —C(O)N(R 2 ) 2 , —C(O)OR 3 , or C(O)NHC(O)OR 2 , where R 2 is hydrogen or a (C 1 -C 10 ) alkyl group and R 3 is a (C 1 -C 10 ) alkyl group.
  • P comprises the indicated number of contiguous amino acids residues from the CRF1 intracellular loop (i1, i2 or i3) or domain (i4) from which it is derived, the remainder of the peptide, if present, can be selected from:
  • peptide backbone modifications such as, but not limited to, those described in (e) above; retro-inverso peptide linkages; despsipeptide linkages; conformational restrictions; or a combination thereof.
  • P of Formula I can be optionally functionalized at the C-terminus.
  • Functionalized at the C-terminus means that the acid moiety present at the C-terminus is replaced by some other functional group. Suitable functional groups include —C(O)N(R 2 ) 2 , —C(O)OR 3 , or C(O)NHC(O)OR 2 , where R 2 is hydrogen or a (C 1 -C 10 ) alkyl group and R 3 is a (C 1 -C 10 ) alkyl group. Functionalization of the C-terminus can result from the methods used to prepare.
  • Peptidomimetic refers to a compound comprising non-peptidic structural elements in place of a peptide sequence.
  • amino acid includes both a naturally occurring amino acid and a non-natural amino acid.
  • naturally occurring amino acid means a compound represented by the formula NH 2 —CHR—COOH, wherein R is the side chain of a naturally occurring amino acids such as lysine, arginine, serine, tyrosine etc. as shown in the Table below.
  • Non-natural amino acid means an amino acid for which there is no nucleic acid codon.
  • non-natural amino acids include, for example, the D-isomers of the natural ⁇ -amino acids such as D-proline (D-P, D-Pro) as indicated above; natural ⁇ -amino acids with non-natural side chains (e.g.,
  • Aib aminobutyric acid
  • bAib 3-aminoisobutyric acid
  • Nva node
  • ⁇ -Ala Aad (2-aminoadipic acid), bAad (3-aminoadipic acid), Abu (2-aminobutyric acid), Gaba ( ⁇ -aminobutyric acid), Acp (6-aminocaproic acid), Dbu (2,4-diaminobutryic acid), ⁇ -aminopimelic acid, TMSA (trimethylsilyl-Ala), aIle (allo-isoleucine), Nle (norleucine), tert-Leu, Cit (citrulline), Orn (ornithine, O), Dpm (2,2′-diaminopimelic acid), Dpr (2,3-diaminopropionic acid), ⁇ or . ⁇ -Nal, Cha (cyclohexyl-Ala),
  • Unnatural amino acids also include cyclic amino acids; and amino acid analogs, for example, N ⁇ -alkylated amino acids such as MeGly (N ⁇ -methylglycine), EtGly (N ⁇ -ethylglycine) and EtAsn (N ⁇ -ethylasparagine); and amino acids in which the ⁇ -carbon bears two side-chain substituents.
  • N ⁇ -alkylated amino acids such as MeGly (N ⁇ -methylglycine), EtGly (N ⁇ -ethylglycine) and EtAsn (N ⁇ -ethylasparagine)
  • amino acids in which the ⁇ -carbon bears two side-chain substituents are two side-chain substituents.
  • the residues of the unnatural amino acids are what are left behind when the unnatural amino acid becomes part of a peptide sequence as described herein.
  • Amino acid residues are amino acid structures as described above that lack a hydrogen atom of the amino group or the hydroxyl moiety of the carboxyl group or both resulting in the units of a peptide chain being amino-acid residues.
  • D-isomers of the natural amino acids are designated herein with a lower case letter of the corresponding naturally occurring amino acid.
  • d-proline is designated “p” rather than “P” as is used for naturally occurring proline.
  • T of Formula I is a lipohilic tether moiety which imparts lipophilicity to the CRF1 receptor compounds of the invention.
  • the lipophilicity which T imparts can promote penetration of the CRF1 receptor compounds into the cell membrane and tethering of the CRF1 receptor compounds to the cell membrane. As such, the lipophilicity imparted by T can facilitate interaction between the CRF1 receptor compounds of the invention and the cognate receptor.
  • the relative lipophilicity of compounds suitable for use as the lipophilic tether moiety of Formula I can be quantified by measuring the amount of the compound that partitions into an organic solvent layer (membrane-like) vs. an aqueous solvent layer (analogous to the extracellular or cytoplasmic environment).
  • Partition coeff P [compound] octanol /[compound] aqueous ).
  • the partition coefficient is expressed in logarithmic form, as the log P. Compounds with greater lipophilicity have a more positive log P than more hydrophilic compounds and tend to interact more strongly with membrane bilayer
  • Computational programs are also available for calculating the partition coefficient for compounds suitable for use as the lipophilic tether moiety (T).
  • T lipophilic tether moiety
  • the trend in log P can be calculated using, for example, ChemDraw (CambridgeSoft, Inc).
  • T is an optionally substituted (C 6 -C 30 )alkyl, (C 6 -C 30 )alkenyl, (C 6 -C 30 )alkynyl wherein 0-3 carbon atoms are replaced with oxygen, sulfur, nitrogen or a combination thereof.
  • the (C 6 -C 30 )alkyl, (C 6 -C 30 )alkenyl, (C 6 -C 30 )alkynyl are substituted at one or more substitutable carbon atoms with halogen, —CN, —OH, —NH 2 , NO 2 , —NH(C 1 -C 6 )alkyl, —N((C 1 -C 6 )alkyl) 2 , (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkoxy, aryloxy, (C 1 -C 6 )alkoxycarbonyl, —CONH 2 , —OCONH 2 , —NHCONH 2 , —N(C 1 -C 6 )alkylCONH 2 , —N(C 1 -C 6 )alkylCONH 2 ,
  • T is selected from the group consisting of: CH 3 (CH 2 ) 9 OPh-, CH 3 (CH 2 ) 6 C ⁇ C(CH 2 ) 6 , CH 3 (CH 2 ) 11 O(CH 2 ) 3 , CH 3 (CH 2 ) 9 O(CH 2 ) 2 and CH 3 (CH 2 ) 13 .
  • T is selected from the group consisting of: CH 3 (CH 2 ) 16 , CH 3 (CH 2 ) 15 , CH 3 (CH 2 ) 14 , CH 3 (CH 2 ) 13 , CH 3 (CH 2 ) 12 , CH 3 (CH 2 ) 11 , CH 3 (CH 2 ) 10 , CH 3 (CH 2 ) 9 , CH 3 (CH 2 ) 8 , CH 3 (CH 2 ) 9 OPh-, CH 3 (CH 2 ) 6 C ⁇ C(CH 2 ) 6 , CH 3 (CH 2 ) 11 O(CH 2 ) 3 , and CH 3 (CH 2 ) 9 O(CH 2 ) 2 and CH 3 (CH 2 ) 13 .
  • the lipophilic moiety (T) of Formula I can be derived from precursor liphophilic compounds (e.g., fatty acids and bile acids).
  • precursor liphophilic compounds e.g., fatty acids and bile acids.
  • “derived from” with regard to T means that T is derived from a precursor lipophilic compound and that reaction of the precursor lipophilic compound in preparing the CRF1 receptor compounds of Formula I, results in a lipophilic tether moiety represented by T in Formula I that is structurally modified in comparison to the precursor lipophilic compound.
  • the lipophilic tether moiety, T of Formula I can be derived from a fatty acid or a bile acid. It is understood that in accordance with Formula I, when T is derived from a fatty acid (i.e., a fatty acid derivative) it is attached to L-P at the carbon atom alpha to the carbonyl carbon of the acid functional group in the fatty acid from which it is derived. For example, when T is derived from palmitic acid,
  • T of Formula I has the following structure:
  • T of Formula I has the following structure:
  • T of Formula I has the following structure:
  • T of Formula I has the following structure:
  • T of Formula I has the following structure:
  • T of Formula I has the following structure:
  • T is derived from 16-hydroxypalmitic acid
  • T of Formula I has the following structure:
  • T is derived from 2-aminooctadecanoic acid
  • T of Formula I has the following structure:
  • T is derived from 2-amino-4-(dodecyloxy)butanoic acid
  • T of Formula I has the following structure:
  • T is derived from a fatty acid.
  • T is derived from a fatty acid selected from the group consisting of: butyric acid, caproic acid, caprylic acid, capric acid, lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, and lignoceric acid.
  • T is derived from a fatty acid selected from the group consisting of: myristoleic acid, palmitoleic acid, oleic acid, linoleic acid, ⁇ -linolenic acid, arachidonic acid, eicosapentaenoic acid, erucic acid, docosahexaenoic acid
  • T of Formula I can be derived from a bile acid. Similar to the embodiment where T is a fatty acid derivative, it is understood that in accordance with Formula I, when T is derived from a bile acid (i.e., a bile acid derivative) it is attached to L-P at the carbon atom alpha to the carbonyl carbon of the acid functional group in the bile acid from which it is derived. For example, when T is derived from lithocholic acid,
  • T of Formula I has the following structure:
  • T is derived from a bile acid.
  • T is derived from a bile acid selected from the group consisting of: lithocholic acid, chenodeoxycholic acid, deoxycholic acid, cholanic acid, cholic acid, ursocholic acid, ursodeoxycholic acid, isoursodeoxycholic acid, lagodeoxycholic acid, dehydrocholic acid, hyocholic acid, hyodeoxycholic acid and the like.
  • T is selected from:
  • T is derived from a bile acid described above that has been modified at other than the acid functional group.
  • T can be derived from any of the bile acids described above, where the hydroxy position has been modified to form an ester or a halo ester.
  • T can be:
  • lipophilic moieties suitable for use as the lipophilic membrane tether, T, of Formula I include but are not limited to steroids.
  • Suitable steroids include, but are not limited to, sterols; progestagens; glucocorticoids; mineralcorticoids; androgens; and estrogens.
  • any steroid capable of attachment or which can be modified for incorporation into Formula I can be used.
  • the lipophilic membrane tether, T may be slightly modified from the precursor lipophilic compound as a result of incorporation into Formula I.
  • Suitable sterols for use in the invention at T include but are not limited to: cholestanol, coprostanol, cholesterol, epicholesterol, ergosterol, ergocalciferol, and the like.
  • Preferred sterols are those that provide a balance of lipophilicity with water solubility.
  • Suitable progestagens include, but are not limited to progesterone.
  • Suitable glucocorticoids include, but are not limited to cortisol.
  • Suitable mineralcorticoids include, but are not limited to aldosterone.
  • Suitable androgens include, but are not limited to testosterone and androstenedione.
  • Suitable estrogens include, but are not limited to estrone and estradiol.
  • T can be derived from 2-tetradecanamideooctadecanoid acid. Similar to the embodiment where T is a fatty acid derivative, it is understood that in accordance with Formula I, when T is derived from 2-tetradecanamideooctadecanoid acid it is attached to L-P at the carbon atom alpha to the carbonyl carbon of the acid functional group in the bile acid from which it is derived. For example, when T is derived from 2-tetradecanamideooctadecanoid acid, the tether is:
  • T of Formula I can be derived from 2-(5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamido)octadecanoic acid.
  • T is derived from 2-(5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamido)octadecanoic acid
  • the tether is:
  • T of Formula I can be:
  • the compounds can contain one of more tether moieties.
  • the tether moieties are the same. In other embodiments, the tether moieties are different.
  • the GPCR Compound of the invention is represented by Formula I:
  • P comprises at least six contiguous amino acid residues.
  • P comprises at least 3 contiguous amino acids of the i1 loop.
  • P is derived from the i1 loop and is represented by the following structural formula or pharmaceutically acceptable salts thereof
  • At least four of X 7 , X 8 , X 9 , X 10 , X 11 , X 12 , X 13 and X 14 are present.
  • X 7 is an arginine residue
  • X 8 is a serine residue
  • X 9 is an isoleucine residue
  • X 10 is an arginine residue.
  • X 11 is a cysteine or serine residue
  • X 12 is a leucine residue
  • X 13 is an arginine residue
  • X 14 is an asparagine residue.
  • X 11 is a cysteine residue.
  • X 11 is a serine residue.
  • X 14 is an asparagine residue
  • X 15 is an isoleucine residue
  • X 16 is an isoleucine residue
  • X 17 is a histidine residue.
  • the i1 loop of the CRF1 receptor from which P is derived has the following sequence: VLFLRLRSIRCLRNIIHWN (SEQ ID NO: 1).
  • P is a sequence selected from:
  • VLFLRLRSIRCLRNIIHWN (SEQ ID NO: 1) VLFLRLRSIRCLRNIIHW; (SEQ ID NO: 2) GSGRLRSIRSLRNIIHWN; (SEQ ID NO: 3) VLFLRLRSIRCLRNIIH; (SEQ ID NO: 4) FLRLRSIRCLRNIIHWN; (SEQ ID NO: 5) GSGRLRSIRSLRNIIH; (SEQ ID NO: 6) LFIRIRSIRSLRNIIH; (SEQ ID NO: 7) VLFLRLRSIRCLRNII; (SEQ ID NO: 8) RLRSIRCLRNIIHWN; (SEQ ID NO: 9) RLRSIRSLRNIIHWN; (SEQ ID NO: 10) VLFLRLRSIRCLRNI; (SEQ ID NO: 11) VLFLRLRSIRCLRN; (SEQ ID NO: 12) RLRSIRSLRNIIH; (SEQ ID NO: 13) RSIRSLRNIIH
  • P comprises at least 3 contiguous amino acids of the i2 loop.
  • P derived from the i2 loop and is represented by the following structural formula or a pharmaceutically acceptable salt thereof
  • At least three of Y 7 , Y 8 , Y 9 , Y 10 , Y 11 , Y 12 , Y 13 , Y 14 , Y 15 , Y 16 , Y 17 , Y 18 and Y 19 are present.
  • Y 9 is serine, threonine or glycine residue
  • Y 10 is a threonine, serine, tyrosine, glycine or alanine residue
  • Y 11 is an aspartic acid, alanine, serine or glycine residue.
  • Y 12 is an arginine or alanine residue
  • Y 13 is a leucine or an alanine residue
  • Y 14 is an arginine or alanine residue.
  • Y 9 is serine, threonine or glycine residue
  • Y 10 is a threonine, serine, tyrosine, glycine or alanine residue
  • Y 11 is an aspartic acid, alanine, serine or glycine residue
  • Y 12 is an arginine or alanine residue.
  • Y 11 is an aspartic acid, alanine, serine or glycine residue
  • Y 12 is an arginine or alanine residue
  • Y 13 is a leucine or an alanine residue
  • Y 14 is an arginine or alanine residue.
  • Y 9 is serine, threonine or glycine residue
  • Y 10 is a threonine, serine, tyrosine, glycine or alanine residue
  • Y 11 is an aspartic acid, alanine, serine or glycine residue
  • Y 12 is an arginine or alanine residue
  • Y 13 is a leucine or an alanine residue.
  • Y 10 is a threonine, serine, tyrosine, glycine or alanine residue
  • Y 11 is an aspartic acid, alanine, serine or glycine residue
  • Y 12 is an arginine or alanine residue
  • Y 13 is a leucine or an alanine residue
  • Y 14 is an arginine or alanine residue.
  • Y 9 is serine, threonine or glycine residue
  • Y 10 is a threonine, serine, tyrosine, glycine or alanine residue
  • Y 11 is an aspartic acid, alanine, serine or glycine residue
  • Y 12 is an arginine or alanine residue
  • Y 13 is a leucine or an alanine residue
  • Y 14 is an arginine or alanine residue.
  • Y 9 is serine residue
  • Y 10 is a threonine residue
  • Y 11 is an aspartic acid residue
  • Y 12 is an arginine residue
  • Y 13 is a leucine residue
  • Y 14 is an arginine residue.
  • Y 7 is a threonine or serine residue
  • Y 8 is a tyrosine or glycine residue
  • Y 9 is a serine, threonine or glycine residue
  • Y 10 is a threonine, serine, tyrosine, glycine or alanine residue.
  • Y 7 is a threonine residue
  • Y 8 is a tyrosine residue
  • Y 9 is a serine residue
  • Y 10 is a threonine residue.
  • Y 11 is an aspartic acid, alanine, serine or glycine residue. Specifically, Y 11 is an aspartic acid residue.
  • Y 12 is an arginine or alanine residue
  • Y 13 is or a leucine or an alanine residue
  • Y 14 is an arginine or alanine residue
  • Y 15 is a lysine or an alanine residue.
  • one of Y 12 , Y 13 , Y 14 , and Y 15 is an alanine residue, provided that the other three of Y 12 , Y 13 , Y 14 , and Y 15 are not an alanine residue.
  • Y 12 is an arginine residue
  • Y 13 is a leucine residue
  • Y 14 is an arginine residue
  • Y 15 is a lysine residue.
  • Y 16 is a tryptophan, methionine or an alanine residue. Specifically, Y 16 is a tryptophan residue. Alternatively, Y 16 is a methionine residue.
  • Y 15 is a lysine or an alanine residue
  • Y 16 is a tryptophan, methionine or an alanine residue
  • Y 17 is a methionine, norleucine, phenylalanine or an alanine residue
  • Y 18 is a phenylalanine, isoleucine or an alanine residue.
  • one of Y 15 , Y 16 , Y 17 , and Y 18 is an alanine residue, provided that the other three of Y 15 , Y 16 , Y 17 , and Y 18 are not an alanine residue.
  • Y 15 is a lysine residue
  • Y 16 is a tryptophan or methionine residue
  • Y 17 is a methionine, norleucine, or phenylalanine residue
  • Y 18 is a phenylalanine or isoleucine residue.
  • Y 15 is a lysine residue
  • Y 16 is a tryptophan residue
  • Y 17 is a methionine residue
  • Y 18 is a phenylalanine residue.
  • Y 19 is an isoleucine or alanine residue. Specifically, Y 19 is an isoleucine residue.
  • the i2 loop of the CRF1 receptor from which P is derived has the following sequence: HTAIVLTYSTDRLRKWMFI (SEQ ID NO: 24).
  • P is a sequence selected from:
  • HTAIVLTYSTDRLRKWMFI HTAIVLTYSTDRLRKWMFI; (SEQ ID NO: 24) HTAIVLTYSTDRLRKWM; (SEQ ID NO: 25) AIVLTYSTDRLRKWMFI; (SEQ ID NO: 26) GSGTYSTDRLRKWMFI; (SEQ ID NO: 27) HTAIVLTYSTDRLRK; (SEQ ID NO: 28) GSGTYSTDRLRKWMF; (SEQ ID NO: 29) VLTYSTDRLRKWMFI; (SEQ ID NO: 30) HTAIVLTYSTDRLR; (SEQ ID NO: 31) LTYSTDRLRKWMFI; (SEQ ID NO: 32) TYSTDRLRKWMFI; (SEQ ID NO: 33) HTAIVLTYSTDR; (SEQ ID NO: 34) GSGTYDRLRKWM; (SEQ ID NO: 35) GSGTYSTDRLRK; (SEQ ID NO: 36) GSTDRLRKWMFI; (SEQ ID NO: 37)
  • P comprises at least 3 contiguous amino acids of the i3 loop.
  • P is derived from the i3 loop and is represented by the following structural formula or a pharmaceutically acceptable salt thereof:
  • Z 1 is absent or a phenylalanine residue
  • Z 2 is absent or an asparagine residue
  • Z 3 is absent or an isoleucine residue
  • Z 4 is absent or a valine residue
  • Z 5 is absent or an arginine residue
  • Z 6 is absent or an isoleucine residue
  • Z 7 is absent or a leucine residue
  • Z 8 is absent or a methionine residue
  • Z 9 is absent or a threonine residue
  • Z 10 is absent or a lysine residue
  • Z 11 is absent or a leucine residue
  • Z 12 is absent or an arginine residue
  • Z 13 is absent or an alanine residue
  • Z 14 is absent or a serine residue
  • Z 15 is absent or a threonine residue
  • Z 16 is absent or a threonine residue
  • Z 17 is absent or a serine residue
  • Z 18 is absent or a glutamic acid residue
  • Z 19 is absent or a threonine residue
  • Z 20 is absent or an isoleucine residue
  • Z 21 is absent or a glutamine residue
  • Z 22 is absent or a tyrosine residue
  • Z 23 is absent or an arginine residue
  • Z 24 is absent or a lysine residue
  • Z 25 is absent or an alanine residue
  • Z 26 is absent or a valine residue
  • Z 27 is absent or a lysine residue
  • Z 28 is absent or an alanine or serine residue
  • Z 29 is absent or a threonine residue
  • Z 30 is absent or a leucine residue
  • R 1 is OR 2 or N(R 2 ) 2 ;
  • each R 2 is independently hydrogen or (C 1 -C 10 )alkyl
  • At least three of Z 5 , Z 6 , Z 7 , Z 8 , and Z 9 are present.
  • Z 5 is an arginine residue
  • Z 6 is an isoleucine residue
  • Z 7 is a leucine residue.
  • Z 7 is a leucine residue
  • Z 8 is a methionine residue
  • Z 9 is a threonine residue.
  • Z 5 is an arginine residue
  • Z 6 is an isoleucine residue
  • Z 7 is a leucine residue
  • Z 8 is a methionine residue.
  • Z 6 is an isoleucine residue
  • Z 7 is a leucine residue
  • Z 8 is absent or a methionine residue
  • Z 9 is a threonine residue.
  • Z 5 is an arginine residue
  • Z 6 is an isoleucine residue
  • Z 7 is a leucine residue
  • Z 8 is a methionine residue
  • Z 9 is a threonine residue.
  • At least three of Z 10 , Z 11 , Z 12 , and Z 13 are present.
  • Z 10 is a lysine residue
  • Z 11 is a leucine residue
  • Z 12 is an arginine residue.
  • Z 11 is a leucine residue
  • Z 12 is an arginine residue
  • Z 13 is an alanine residue.
  • Z 10 is a lysine residue
  • Z 11 is a leucine residue
  • Z 12 is an arginine residue
  • Z 13 is an alanine residue.
  • At least three of Z 14 , Z 15 , Z 16 , and Z 17 are present.
  • Z 14 is a serine residue
  • Z 15 is a threonine residue
  • Z 16 is a threonine residue.
  • Z 15 is a threonine residue
  • Z 16 is a threonine residue
  • Z 17 is a serine residue
  • Z 14 is a serine residue
  • Z 15 is a threonine residue
  • Z 16 is a threonine residue
  • Z 17 is a serine residue
  • At least three of Z 19 , Z 20 , Z 21 , Z 22 , and Z 23 are present.
  • Z 19 is a threonine residue
  • Z 20 is an isoleucine residue
  • Z 21 is a glutamine residue.
  • Z 21 is a glutamine residue
  • Z 22 is a tyrosine residue
  • Z 23 is an arginine residue.
  • Z 19 is a threonine residue
  • Z 20 is an isoleucine residue
  • Z 21 is a glutamine residue
  • Z 22 is a tyrosine residue.
  • Z 20 is an isoleucine residue
  • Z 21 is a glutamine residue
  • Z 22 is a tyrosine residue
  • Z 23 is an arginine residue.
  • Z 19 is a threonine residue
  • Z 20 is an isoleucine residue
  • Z 21 is a glutamine residue
  • Z 22 is a tyrosine residue
  • Z 23 is an arginine residue.
  • At least three of Z 24 , Z 25 , Z 26 , Z 27 , and Z 28 are present.
  • Z 24 is a lysine residue
  • Z 25 is an alanine residue
  • Z 26 is a valine residue.
  • Z 26 is a valine residue
  • Z 27 is a lysine residue
  • Z 28 is an alanine or serine residue.
  • Z 24 is a lysine residue
  • Z 25 is an alanine residue
  • Z 26 is a valine residue
  • Z 27 is a lysine residue.
  • Z 25 is an alanine residue
  • Z 26 is a valine residue
  • Z 27 is a lysine residue
  • Z 28 is an alanine or serine residue.
  • Z 24 is a lysine residue
  • Z 25 is an alanine residue
  • Z 26 is a valine residue
  • Z 27 is a lysine residue
  • Z 28 is an alanine or serine residue. Specifically, Z 28 is an alanine residue.
  • the i3 loop of the CRF1 receptor from which P is derived has the following sequence: FNIVRILMTKLRASTTSETIQYRKAVKA (SEQ ID NO: 71).
  • P is a sequence selected from:
  • FNIVRILMTKLRASTTSETIQYRKAVKA (SEQ ID NO: 71) IVRILMTKLRASTTSETIQYRKAVKA; (SEQ ID NO: 72) FNIVRILMTKLRASTTSETIQYRKAV; (SEQ ID NO: 73) RILMTKLRASTTSETIQYRKAVKA; (SEQ ID NO: 74) FNIVRILMTKLRASTTSETIQYRK; (SEQ ID NO: 75) FNIVRILMTKLRASTTSETIQYR; (SEQ ID NO: 76) LMTKLRASTTSETIQYRKAVKA; (SEQ ID NO: 77) FNIVRILMTKLRASTTSETIQY; (SEQ ID NO: 78) TKLRASTTSETIQYRKAVKA; (SEQ ID NO: 79) LMTKLRASTTSETIQYRKAV; (SEQ ID NO: 80) LRASTTSETIQYRKAVKA; (SEQ ID NO: 81) LM
  • P comprises at least 3 contiguous amino acids of the i4 domain.
  • P derived from the i4 domain and is represented by the following structural formula or a pharmaceutically acceptable salt thereof,
  • M 1 is absent or a glutamic acid residue
  • M 2 is absent or a valine residue
  • M 3 is absent or an arginine residue
  • M 4 is absent or a serine residue
  • M 5 is absent or an alanine residue
  • M 6 is absent or an isoleucine residue
  • M 7 is absent or an arginine residue
  • M 8 is absent or a lysine residue
  • M 9 is absent or an arginine residue
  • M 10 is absent or a tryptophan residue
  • M 11 is absent or a histidine residue
  • M 12 is absent or an arginine residue
  • M 13 is absent or a tryptophan residue
  • M 14 is absent or a glutamine residue
  • M 15 is absent or an aspartic acid residue
  • M 16 is absent or a lysine residue
  • M 17 is absent or a histidine or glycine residue
  • M 18 is absent or a serine residue
  • M 19 is absent or an isoleucine residue
  • M 20 is absent or an arginine residue
  • M 21 is absent or an alanine residue
  • M 22 is absent or an arginine residue
  • M 23 is absent or a valine residue
  • M 24 is absent or an alanine residue
  • M 25 is absent or an arginine residue
  • M 26 is absent or an alanine residue
  • M 27 is absent or a methionine residue
  • M 28 is absent or a serine residue
  • M 29 is absent or an isoleucine residue
  • M 30 is absent or a proline residue
  • M 31 is absent or a threonine residue
  • M 32 is absent or a serine residue
  • M 33 is absent or a proline residue
  • M 34 is absent or a threonine residue
  • M 35 is absent or an arginine residue
  • M 36 is absent or a valine residue
  • M 37 is absent or a serine residue
  • M 38 is absent or a phenylalanine residue
  • M 39 is absent or a histidine residue
  • M 40 is absent or a serine residue
  • M 41 is absent or an isoleucine residue
  • M 42 is absent or a lysine residue
  • M 43 is absent or a glutamine residue
  • M 44 is absent or a serine residue
  • M 45 is absent or a threonine residue
  • M 46 is absent or an alanine residue
  • M 47 is absent or a valine residue; provided that at least five of M 1 -M 47 are present;
  • R 1 is OR 2 or N(R 2 ) 2 ;
  • each R 2 is independently hydrogen or (C 1 -C 10 )alkyl
  • M 1 -M 47 it is bonded to L as written. For example, M 1 is bonded to L. If M 1 is absent, then M 2 is bonded to L.
  • At least three of M 2 -M 7 are present.
  • M 2 is a valine residue
  • M 3 is an arginine residue
  • M 4 is a serine residue.
  • M 5 is an alanine residue
  • M 6 is an isoleucine residue
  • M 7 is an arginine residue.
  • M 2 is a valine residue
  • M 3 is an arginine residue
  • M 4 is a serine residue
  • M 5 is an alanine residue.
  • M 4 is a serine residue
  • M 5 is an alanine residue
  • M 6 is an isoleucine residue
  • M 7 is an arginine residue.
  • M 2 is a valine residue
  • M 3 is an arginine residue
  • M 4 is a serine residue
  • M 5 is an alanine residue
  • M 6 is an isoleucine residue.
  • M 3 is an arginine residue
  • M 4 is a serine residue
  • M 5 is an alanine residue
  • M 6 is an isoleucine residue
  • M 7 is an arginine residue.
  • M 2 is a valine residue
  • M 3 is an arginine residue
  • M 4 is a serine residue
  • M 5 is an alanine residue
  • M 6 is an isoleucine residue
  • M 7 is an arginine residue.
  • At least three of M 9 -M 13 are present.
  • M 9 is an arginine residue
  • M 10 is a tryptophan residue
  • M 11 is a histidine residue
  • M 12 is an arginine residue.
  • M 10 is a tryptophan residue
  • M 11 is a histidine residue
  • M 12 is an arginine residue
  • M 13 is a tryptophan residue.
  • M 9 is an arginine residue
  • M 10 is a tryptophan residue
  • M 11 is a histidine residue
  • M 12 is an arginine residue
  • M 13 is a tryptophan residue.
  • At least three of M 18 -M 22 are present.
  • M 18 is a serine residue
  • M 19 is an isoleucine residue
  • M 20 is an arginine residue
  • M 21 is an alanine residue.
  • M 19 is an isoleucine residue
  • M 20 is an arginine residue
  • M 21 is an alanine residue
  • M 22 is an arginine residue.
  • M 18 is a serine residue
  • M 19 is an isoleucine residue
  • M 20 is an arginine residue
  • M 21 is an alanine residue
  • M 22 is an arginine residue.
  • At least three of M 23 -M 27 are present.
  • M 23 is a valine residue
  • M 24 is an alanine residue
  • M 25 is an arginine residue
  • M 26 is an alanine residue.
  • M 24 is an alanine residue
  • M 25 is an arginine residue
  • M 26 is an alanine residue
  • M 27 is a methionine residue.
  • M 23 is a valine residue
  • M 24 is an alanine residue
  • M 25 is an arginine residue
  • M 26 is an alanine residue
  • M 27 is a methionine residue.
  • At least three of M 25 -M 29 are present.
  • M 25 is an arginine residue
  • M 26 is an alanine residue
  • M 27 is a methionine residue
  • M 28 is a serine residue.
  • M 26 is an alanine residue
  • M 27 is a methionine residue
  • M 28 is a serine residue
  • M 29 is an isoleucine residue.
  • M 25 is an arginine residue
  • M 26 is an alanine residue
  • M 27 is a methionine residue
  • M 28 is a serine residue
  • M 29 is an isoleucine residue.
  • At least three of M 32 -M 36 are present.
  • M 32 is a serine residue
  • M 33 is a proline residue
  • M 34 is a threonine residue
  • M 35 is an arginine residue.
  • M 33 is a proline residue
  • M 34 is a threonine residue
  • M 35 is an arginine residue
  • M 36 is a valine residue.
  • M 32 is a serine residue
  • M 33 is a proline residue
  • M 34 is a threonine residue
  • M 35 is an arginine residue
  • M 36 is a valine residue.
  • the i4 domain of the CRF1 receptor from which P is derived has the following sequence:
  • P is a sequence selected from:
  • RAMSIPTSPTRVSFHSIKQSTAV (SEQ ID NO: 100) HSIRARVARAMSIPTSPTRV; (SEQ ID NO: 101) RWQDKHSIRARVARAMSI; (SEQ ID NO: 102) EVRSAIRKRWHRWQD; (SEQ ID NO: 103) RWHRWQDKHSIRAR; (SEQ ID NO: 104) VRSAIRKRWHRW; (SEQ ID NO: 105) and GSIRARVARAM. (SEQ ID NO: 106)
  • T is an optionally substituted (C 6 -C 30 )alkyl, (C 6 -C 30 )alkenyl, (C 6 -C 30 )alkynyl, wherein 0-3 carbon atoms are replaced with oxygen, sulfur, nitrogen or a combination thereof.
  • This value of T is applicable to the first, second, third, fourth, fifth and sixth aspects and the specific (i.e., specific, more specific and most specific) embodiments of same.
  • T is selected from: CH 3 (CH 2 ) 16 , CH 3 (CH 2 ) 15 , CH 3 (CH 2 ) 14 , CH 3 (CH 2 ) 13 , CH 3 (CH 2 ) 12 , CH 3 (CH 2 ) 11 , CH 3 (CH 2 ) 10 , CH 3 (CH 2 ) 9 , CH 3 (CH 2 ) 8 , CH 3 (CH 2 ) 9 OPh-, CH 3 (CH 2 ) 6 C ⁇ C(CH 2 ) 6 , CH 3 (CH 2 ) 11 O(CH 2 ) 3 , and CH 3 (CH 2 ) 9 O(CH 2 ) 2 .
  • T is a fatty acid derivative.
  • the fatty acid is selected from the group consisting of: butyric acid, caproic acid, caprylic acid, capric acid, lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, lignoceric acid, myristoleic acid, palmitoleic acid, oleic acid, linoleic acid, ⁇ -linolenic acid, arachidonic acid, eicosapentaenoic acid, erucic acid, docosahexaenoic acid.
  • T is a bile acid derivative. This value of T is applicable to the first, second, third, fourth, fifth and sixth aspects and the specific (i.e., specific, more specific and most specific) embodiments of same.
  • the bile acid is selected from the group consisting of: lithocholic acid, chenodeoxycholic acid, deoxycholic acid, cholanic acid, cholic acid, ursocholic acid, ursodeoxycholic acid, isoursodeoxycholic acid, lagodeoxycholic acid, dehydrocholic acid, hyocholic acid, and hyodeoxycholic acid.
  • T is selected from sterols; progestagens; glucocorticoids; mineralcorticoids; androgens; and estrogens. This value of T is applicable to the first, second, third, fourth, fifth and sixth aspects and the specific (i.e., specific, more specific and most specific) embodiments of same.
  • T-L of Formula I is represented by a moiety selected from the group consisting of:

Abstract

The invention relates generally to compounds which are allosteric modulators (e.g., negative and positive allosteric modulators, allosteric agonists, and ago-allosteric modulators) of the G protein coupled receptor for corticotrophin releasing hormone (or factor) receptor 1, also known as the CRF1, CRHR1, CRFR1, CRHR, CRF-R. The CRF1 receptor compounds are derived from the intracellular loops and domains of CRF1 receptor. The invention also relates to the use of these CRF1 receptor compounds and pharmaceutical compositions comprising the CRF1 receptor compounds in the treatment of diseases and conditions associated with CRF1 receptor modulation, such as inflammatory bowel disease (peripherally acting), irritable bowel syndrome (IBS), stress response (colonic motor activity), anxiety, sleep disorder, addictive behavior, acute and chronic neurodegeneration, preterm labor and pain.

Description

    RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Application No. 61/198,194, filed on Nov. 4, 2008. The entire teachings of the above application is incorporated herein by reference.
  • BACKGROUND OF THE INVENTION
  • G protein coupled receptors (GPCRs) constitute one of the largest families of genes in the human genome. GPCRs are integral membrane signaling proteins. Hydrophobicity mapping of the amino acid sequences of G-protein coupled receptors has led to a model of the typical G-protein-coupled receptor as containing seven hydrophobic membrane-spanning regions with the amino terminal on the extracellular side of the membrane and the carboxyl terminal on the intracellular side of the membrane.
  • GPCRs mediate the transmission of intracellular signals (“signal transduction”) by activating guanine nucleotide-binding proteins (G proteins) to which the receptor is coupled. GPCRs are activated by a wide range of endogenous stimuli, including peptides, amino acids. hormones, light, and metal ions. The following reviews are incorporated by reference: Hill, British J. Pharm 147: s27 (2006); Palczeski, Ann Rev Biochemistry 75: 743-767 (2006); Dorsham & Gutkind, Nature Reviews 7: 79-94 (2007); Kobilka & Schertler, Trends Pharmacol Sci. 2: 79-83 (2008).
  • GPCRs are important targets for drug discovery as they are involved in a wide range of cellular signaling pathways and are implicated in many pathological conditions (e.g., cardiovascular and mental disorders, cancer, AIDS). In fact, GPCRs are targeted by 40-50% of approved drugs, illustrating the critical importance of this class of pharmaceutical targets. Interestingly, this number represents only about 30 GPCRs, a small fraction of the total number of GPCRs thought to be relevant to human disease. Over 1000 GPCRs are known in the human genome, and GPCRs remain challenging targets from a research and development perspective in part because these amembrane bound receptors with complex pharmacology.
  • There remains a need for the development of new pharmaceuticals that are GPCR modulators (e.g., agonists, partial agonists, inverse agonists and antagonists) and especially those that are allosteric modulators of GPCRs (e.g., negative and positive allosteric modulators, allosteric agonists, and ago-allosteric modulators).
  • SUMMARY OF THE INVENTION
  • The invention relates generally to compounds which are allosteric modulators (e.g., negative and positive allosteric modulators, allosteric agonists, and ago-allosteric modulators) of the G protein coupled receptor for corticotrophin releasing hormone (or factor) receptor 1, also known as the CRF1, CRHR1, CRFR1, CRHR, CRF-R. The CRF1 receptor compounds are derived from the intracellular loops and domains of CRF1 receptor. The invention also relates to the use of these CRF1 receptor compounds and pharmaceutical compositions comprising the CRF1 receptor compounds in the treatment of diseases and conditions associated with CRF1 receptor modulation, such as inflammatory bowel disease (peripherally acting), irritable bowel syndrome (IBS), stress response (colonic motor activity), anxiety, sleep disorder, addictive behavior, acute and chronic neurodegeneration, preterm labor and pain.
  • More specifically, the invention relates to a compound represented by Formula I:

  • T-L-P,
  • or pharmaceutically acceptable salts thereof, wherein:
      • P is a peptide comprising at least three contiguous amino-acid residues of an intracellular i1, i2, i3 loop or an intracellular i4 domain of the CRF1 receptor;
      • L is a linking moiety represented by C(O) and bonded to P at an N terminal nitrogen of an N-terminal amino-acid residue;
      • and T is a lipophilic tether moiety bonded to L, wherein the C-terminal amino acid residue of P is optionally functionalized.
  • The invention also relates to pharmaceutical compositions comprising one or more compounds of the invention and a carrier, and the use of the disclosed compounds and compositions in methods of treating diseases and conditions responsive to modulation of the CRF1 receptor.
  • DETAILED DESCRIPTION OF THE INVENTION
  • A description of example embodiments of the invention follows.
  • G Protein Coupled Receptors (GPCRs)
  • G protein coupled receptors (GPCRs) constitute one of the largest superfamilies of genes in the human genome; these transmembrane proteins enable the cell the respond to its environment by sensing extracellular stimuli and initiating intracellular signal transduction cascades. GPCRs mediate signal transduction through the binding and activation of guanine nucleotide-binding proteins (G proteins) to which the receptor is coupled. Wide arrays of ligands bind to these receptors, which in turn orchestrate signaling networks integral to many cellular functions. Diverse GPCR ligands include small proteins, peptides, amino acids, biogenic amines, lipids, ions, odorants and even photons of light. The following reviews are incorporated by reference: Hill, British J. Pharm 147: s27 (2006); Dorsam & Gutkind, Nature Reviews 7: 79-94 (2007).
  • In addition to modulating a diverse array of homeostatic processes, GPCR signaling pathways are integral components of many pathological conditions (e.g., cardiovascular and mental disorders, cancer, AIDS). In fact, GPCRs are targeted by 40-50% of approved drugs illustrating the critical importance of this class of pharmaceutical targets. Interestingly, this number represents only about 30 GPCRs, a small fraction of the total number of GPCRs thought to be relevant to human disease. GPCRs are membrane bound receptors that exhibit complex pharmacological properties and remain challenging targets from a research and development perspective. Given their importance in human health combined with their prevalence (over 1000 known GPCRs in the human genome) GPCRs represent an important target receptor class for drug discovery and design.
  • GPCRs are integral membrane proteins that mediate diverse signaling cascades through an evolutionarily conserved structural motif. All GPCRs are thought to consist of seven hydrophobic transmembrane spanning α-helices with the amino terminus on the extracellular side of the membrane and the carboxyl terminus on the intracellular side of the membrane. The transmembrane helices are linked together sequentially by extracellular (e1, e2, e3) and intracellular (cytoplasmic) loops (i1, i2, i3). The intracellular loops or domains are intimately involved in the coupling and turnover of G proteins and include: i1, which connects TM1-TM2; i2, connecting TM3-TM4; i3, connecting TM5-TM6; and a portion of the C-terminal cytoplasmic tail (domain 4). Due in part to the topological homology of the 7™ domains and the recent high resolution crystal structures of several GPCRs (Palczewski et al., Science 289, 739-45 (2000), Rasmussen, S. G. et al., Nature 450, 383-7 (2007)) skilled modelers are now able to predict the general boundaries of GPCR loop domains through the alignment of several related receptors. These predictions are aided in part by a number of programs used by computational biologists, including EMBOSS, ClustalW2, Kalign, and MAFFT (Multiple Alignment using Fast Fourier Transform). Importantly, many of these programs are publically available (see, for example, The European Bioinformatics Institute (EMBL-EBI) web site http://www.ebi.ac.uk/Tools/) and most have web-based interfaces.
  • GPCR mediated signal transduction is initiated by the binding of a ligand to its cognate receptor. In many instances GPCR ligand binding is believed to take place in a hydrophilic pocket generated by a cluster of helices near the extracellular domain. However, other ligands, such as large peptides, are thought to bind to the extracellular region of protein and hydrophobic ligands are postulated to intercalate into a receptor binding pocket through the membrane between gaps in the helices. The process of ligand binding induces conformational changes within the receptor. These changes involve the outward movement of helix 6, which in turn alters the conformations of the intracellular loops and ultimately results in a receptor form that is able to bind and activate a heterotrimeric G protein (Farrens, D., et al. Science 274, 768-770 (1996), Gether, U. and Kobilka, B., J. Biol. Chem. 273, 17979-17982 (1998)). Upon binding the receptor catalyzes the exchange of GTP for GDP in the alpha subunit of the heterotrimeric G protein, which results in a separation of the G protein from the receptor as well a dissociation of the alpha and beta/gamma subunits of the G protein itself. Notably, this process is catalytic and results in signal amplification in that activation of one receptor may elicit the activation and turnover of numerous G proteins, which in turn may regulate multiple second messenger systems. Signaling diversity is further achieved through the existence of numerous G protein types as well as differing isoforms of alpha, beta and gamma subunits. Typically, GPCRs interact with G proteins to regulate the synthesis or inhibition of intracellular second messengers such as cyclic AMP, inositol phosphates, diacylglycerol and calcium ions, thereby triggering a cascade of intracellular events that eventually leads to a biological response.
  • GPCR signaling may be modulated and attenuated through cellular machinery as well as pharmacological intervention. Signal transduction may be ‘switched off’ with relatively fast kinetics (seconds to minutes) by a process called rapid desensitization. For GPCRs, this is caused by a functional uncoupling of receptors from heterotrimeric G proteins, without a detectable change in the total number of receptors present in cells or tissues. This process involves the phosphorylation of the receptor C terminus, which enables the protein arrestin to bind to the receptor and occlude further G protein coupling. Once bound by arrestin the receptor may be internalized into the cell and either recycled back to the cell surface or degraded. The alpha subunit of the G protein possesses intrisic GTPase activity, which attenuates signaling and promotes re-association with the beta/gamma subunits and a return to the basal state. GPCR signaling may also be modulated pharmacologically. Agonist drugs act directly to activate the receptors, whereas antagonist drugs act indirectly to block receptor signaling by preventing agonist activity through their associating with the receptor. GPCR binding and signaling can also be modified through allosteric modulation, that is by ligands that bind not at the orthosteric binding site but through binding at an allosteric site elsewhere in the receptors. Allosteric modulators can include both positive and negative modulators of orthosteric ligand mediated activity, allosteric agonists (that act in the absence of the orthosteric ligand), and ago-allosteric modulators (ligands that have agonist activity on their own but that can also modulate the activity of the orthosteric ligand).
  • The large superfamily of GPCRs may be divided into subclasses based on structural and functional similarities. GPCR families include Class A Rhodopsin like, Class B Secretin like, Class C Metabotropic glutamate/pheromone, Class D Fungal pheromone, Class E cAMP receptors (Dictyostelium), the Frizzled/Smoothened family, and various orphan GPCRs. In addition, putative families include Ocular albinism proteins, Insect odorant receptors, Plant Mlo receptors, Nematode chemoreceptors, Vomeronasal receptors (VIR & V3R) and taste receptors.
  • CRF1 is a class B GPCR, also called family B or secretin-like. In general, class B receptors are activated by peptide ligands typically 30 to 40 amino acids in length. Activation of these receptors results in activation of adenylyl cyclase and signal transduction through increase in cAMP as a primary signaling pathway. Class B receptors have a large N-terminal extracellular domain with 4 very highly conserved cysteine residues. This domain is important for the binding of endogenous peptide ligands and resulting receptor activation. While these receptors signal primarily through Gs activation of adenylyl cyclase, they also couple to Gq, resulting in calcium release and may also couple to Gi/GO, which modulate adenylyl cyclase activity.
  • Expression of CRF1 is widespread in the CNS and found at lower levels in peripheral tissues including skin, ovary, testis and adrenal gland. CRF1 plays a pivotal role in the hypothalamic-pituitary-adrenal axis response to stress. Of the CRF family of peptides, which includes corticotrophin releasing hormone (CRF), urocortin I, II, and III, CRF1 is activated only by CRF and urocortin I. In the gastrointestinal (GI) tract, CRF1 activation mediates colonic motility, while activation of the related receptor CRF2 delays gastric emptying. At least 8 isoforms of CRF1 have been identified, but not all isoforms appear to be functional. CRF1-deficient mice possess an impaired stress response and reduced anxiety.
  • Peptides
  • As defined herein, P is a peptide comprising at least three contiguous amino-acid residues (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18 or 19) of an intracellular i1, i2 or i3 loop or intracellular i4 domain of the CRF1 receptor. It is understood that, the N-terminal nitrogen of the N-terminal amino acid residue of P to which the linking moiety C(O) is bonded can be one of the at least three contiguous amino acid residues or it can be an amino acid residue distinct from the at least three contiguous amino acid residues.
  • Intracellular i1 loop as used herein refers to the loop which connects TM1 to TM2 and the corresponding transmembrane junctional residues.
  • Intracellular i2 loop as used herein refers to the loop which connects TM3 to TM4 and the corresponding transmembrane junctional residues.
  • Intracellular i3 loop as used herein refers to the loop which connects TM5 to TM6 and the corresponding transmembrane junctional residues.
  • Intracellular i4 domain as used herein refers to the C-terminal cytoplasmic tail and the transmembrane junctional residue.
  • In a specific embodiment, P comprises at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, at least fifteen, at least sixteen, at least seventeen, at least eighteen or at least nineteen contiguous amino acid residues of the intracellular i1, i2 or i3 loop or intracellular i4 domain of the CRF1 receptor
  • In a more specific embodiment, the at least three contiguous amino acids of P (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or 19) are derived from the intracellular i1, i2 or i3 loop or intracellular i4 domain of the CRF1 receptor, wherein the amino acid sequence of each loop and the i4 domain is as described in Table 1.
  • TABLE 1
    Intra-
    cellular
    Loop
    or
    Domain CRF1 Receptor
    i1 VLFLRLRSIRCLRNIIHWN (SEQ ID NO: 1)
    i2 HTAIVLTYSTDRLRKWMFI (SEQ ID NO: 24)
    i3 FNIVRILMTKLRASTTSETIQYRKAVKA
    (SEQ ID NO: 71)
    i4 EVRSAIRKRWHRWQDKHSIRARVARAMSIPTSPTRVS
    FHSIKQSTAV (SEQ ID NO: 99)
  • It is understood that in addition to the amino acids in the sequences in Table 1, the intracellular loop for the i1 loop, i2 loop, i3 loop and i4 domain can also include amino acids from transmembrane junctional residues. For example, the i1 loop can include SEQ ID NO: 1 where one or more residues from the transmembrane junctional residues are included on either the C-terminus, the N-terminus or both. For example, SEQ ID NO: 1 can include either an Threonine residue, Leucine residue or both at the C-terminus.
  • In another embodiment, P comprises at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, at least fifteen, at least sixteen, at least seventeen, at least eighteen or at least nineteen contiguous amino acid residues of the i1 intracellular loop of the CRF1 receptor.
  • It is understood that for the embodiments presented herein, that when the amino acid residues of P are represented by X, M, Y or Z that the C-terminal amino acid residue does not include the —OH of the amino acid and that the end group R1 that is bonded to the C-terminal residue includes —OH.
  • In one embodiment, P is derived from the i1 loop and is represented by the following structural formula or pharmaceutically acceptable salts thereof
  • X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11-X12-X13-X14-X15-X16-
    X17-X18-X19-R1
  • wherein:
      • X1 is absent or a valine residue;
      • X2 is absent or a leucine or glycine residue;
      • X3 is absent or a phenylalanine or serine residue;
      • X4 is absent or a leucine or glycine residue;
      • X5 is absent or an arginine residue;
      • X6 is absent or a leucine or isoleucine residue;
      • X7 is absent or an arginine residue;
      • X8 is absent or a serine residue;
      • X9 is absent or an isoleucine residue;
      • X10 is absent or an arginine residue;
      • X11 is absent or a cysteine or serine residue;
      • X12 is absent or a leucine residue;
      • X13 is absent or an arginine residue;
      • X14 is absent or an asparagine residue;
      • X15 is absent or an isoleucine residue;
      • X16 is absent or an isoleucine residue;
      • X17 is absent or a histidine residue;
      • X18 is absent or tryptophan residue;
      • X19 is absent or an asparagine residue; provided that at least five of X1-X19 are present;
      • R1 is OR2 or N(R2)2;
      • each R2 is independently hydrogen or (C1-C10)alkyl; and
      • from 0 to 5 amino acid residues are present in the D configuration.
  • It is understood that when P is described as X1-X19 it is bonded to L as written. For example, X1 is bonded to L. If X1 is absent, then X2 is bonded to L.
  • In a specific embodiment, at least four of X7, X8, X9, X10, X11, X12, X13 and X14 are present.
  • In a further specific embodiment,
  • X7 is an arginine residue;
  • X8 is a serine residue;
  • X9 is an isoleucine residue; and
  • X10 is an arginine residue.
  • In another further specific embodiment,
  • X11 is a cysteine or serine residue;
  • X12 is a leucine residue;
  • X13 is an arginine residue; and
  • X14 is an asparagine residue.
  • In another further specific embodiment, X11 is a cysteine residue. Alternatively, wherein X11 is a serine residue.
  • In another specific embodiment,
  • X14 is an asparagine residue;
  • X15 is an isoleucine residue;
  • X16 is an isoleucine residue; and
  • X17 is a histidine residue.
  • In an even more specific embodiment, P is selected from the group consisting of SEQ ID NOS: 1-23 as listed in Table 2 below:
  • TABLE 2
    SEQ
    i loop CRF1 Sequence ID NO:
    i1     VLFLRLRSIRCLRNIIHWN 1
    i1     VLFLRLRSIRCLRNIIHW 2
    i1      GSGRLRSIRSLRNIIHWN 3
    i1     VLFLRLRSIRCLRNIIH 4
    i1       FLRLRSIRCLRNIIHWN 5
    i1      GSGRLRSIRSLRNIIH 6
    i1      LFIRIRSIRSLRNIIH 7
    i1     VLFLRLRSIRCLRNII 8
    i1         RLRSIRCLRNIIHWN 9
    i1         RLRSIRSLRNIIHWN 10
    i1     VLFLRLRSIRCLRNI 11
    i1     VLFLRLRSIRCLRN 12
    i1         RLRSIRSLRNIIH 13
    i1           RSIRSLRNIIHWN 14
    i1     VLFLRLRSIRCLR 15
    i1           RSIRCLRNIIHW 16
    i1           RSIRSLRNIIH 17
    i1           RSIRCLRNIIH 18
    i1         RLRSIRSLRN 19
    i1              RSLRNIIHWN 20
    i1         RLRSIRSLR 21
    i1              RSLRNIIH 22
    i1           RSIRSLRIT 23
  • In another specific embodiment, the at least three contiguous amino acids of P (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or 19) are derived from the i2 intracellular loop of the CRF1 receptor.
  • In one embodiment, P derived from the i2 loop and is represented by the following structural formula or a pharmaceutically acceptable salt thereof
  • Y1-Y2-Y3-Y4-Y5-Y6-Y7-Y8-Y9-Y10-Y11-Y12-Y13-Y14-Y15-Y16-
    Y17-Y18-Y19-R1,
  • wherein:
      • Y1 is absent or a histidine residue;
      • Y2 is absent or a threonine residue;
      • Y3 is absent or an alanine residue;
      • Y4 is absent or an isoleucine or a glycine residue;
      • Y5 is absent or a valine or serine residue;
      • Y6 is absent or a leucine or glycine residue;
      • Y7 is absent or a threonine or serine residue;
      • Y8 is absent or a tyrosine or glycine residue;
      • Y9 is absent or serine, threonine or glycine residue;
      • Y10 is absent or a threonine, serine, tyrosine, glycine or alanine residue;
      • Y11 is absent or an aspartic acid, alanine, serine or glycine residue;
      • Y12 is absent or an arginine or alanine residue;
      • Y13 is absent or a leucine or an alanine residue;
      • Y14 is absent or an arginine or alanine residue;
      • Y15 is absent or a lysine or an alanine residue;
      • Y16 is absent or a tryptophan, methionine or an alanine residue;
      • Y17 is absent or a methionine, norleucine, phenylalanine or an alanine residue;
      • Y18 is absent or a phenylalanine, isoleucine or an alanine residue;
      • Y19 is absent or an isoleucine or alanine residue, provided that at least five of Y1-Y19, are present; and
      • R1 is OR2 or N(R2)2;
      • each R2 is independently hydrogen or (C1-C10)alkyl; and
      • from 0 to 5 amino acid residues are present in the D configuration.
  • It is understood that when P is described as Y1-Y19 it is bonded to L as written. For example, Y1 is bonded to L. If Y1 is absent, then Y2 is bonded to L.
  • In a specific embodiment, at least three of Y7, Y8, Y9, Y10, Y11, Y12, Y13, Y14, Y15, Y16, Y17, Y18 and Y19 are present.
  • In a further specific embodiment,
  • Y9 is serine, threonine or glycine residue;
  • Y10 is a threonine, serine, tyrosine, glycine or alanine residue; and
  • Y11 is an aspartic acid, alanine, serine or glycine residue.
  • In another further specific embodiment,
  • Y12 is an arginine or alanine residue;
  • Y13 is a leucine or an alanine residue; and
  • Y14 is an arginine or alanine residue.
  • In a further specific embodiment,
  • Y9 is serine, threonine or glycine residue;
  • Y10 is a threonine, serine, tyrosine, glycine or alanine residue;
  • Y11 is an aspartic acid, alanine, serine or glycine residue; and
  • Y12 is an arginine or alanine residue.
  • In a further specific embodiment,
  • Y11 is an aspartic acid, alanine, serine or glycine residue;
  • Y12 is an arginine or alanine residue;
  • Y13 is a leucine or an alanine residue; and
  • Y14 is an arginine or alanine residue.
  • In a further specific embodiment,
  • Y9 is serine, threonine or glycine residue;
  • Y10 is a threonine, serine, tyrosine, glycine or alanine residue;
  • Y11 is an aspartic acid, alanine, serine or glycine residue;
  • Y12 is an arginine or alanine residue; and
  • Y13 is a leucine or an alanine residue.
  • In a further specific embodiment,
  • Y10 is a threonine, serine, tyrosine, glycine or alanine residue;
  • Y11 is an aspartic acid, alanine, serine or glycine residue;
  • Y12 is an arginine or alanine residue;
  • Y13 is a leucine or an alanine residue; and
  • Y14 is an arginine or alanine residue.
  • In yet another further specific embodiment,
  • Y9 is serine, threonine or glycine residue;
  • Y10 is a threonine, serine, tyrosine, glycine or alanine residue;
  • Y11 is an aspartic acid, alanine, serine or glycine residue;
  • Y12 is an arginine or alanine residue;
  • Y13 is a leucine or an alanine residue; and
  • Y14 is an arginine or alanine residue.
  • In yet another further specific embodiment,
  • Y9 is serine residue;
  • Y10 is a threonine residue;
  • Y11 is an aspartic acid residue;
  • Y12 is an arginine residue;
  • Y13 is a leucine residue; and
  • Y14 is an arginine residue.
  • In a specific embodiment,
  • Y7 is a threonine or serine residue;
  • Y8 is a tyrosine or glycine residue;
  • Y9 is a serine, threonine or glycine residue; and
  • Y10 is a threonine, serine, tyrosine, glycine or alanine residue.
  • In another specific embodiment,
  • Y7 is a threonine residue;
  • Y8 is a tyrosine residue;
  • Y9 is a serine residue; and
  • Y10 is a threonine residue.
  • In a further specific embodiment, Y11 is an aspartic acid, alanine, serine or glycine residue. Specifically, Y11 is an aspartic acid residue.
  • In another specific embodiment,
  • Y12 is an arginine or alanine residue;
  • Y13 is or a leucine or an alanine residue;
  • Y14 is an arginine or alanine residue; and
  • Y15 is a lysine or an alanine residue.
  • In another specific embodiment, one of Y12, Y13, Y14, and Y15 is an alanine residue, provided that the other three of Y12, Y13, Y14, and Y15 are not an alanine residue.
  • In another specific embodiment,
  • Y12 is an arginine residue;
  • Y13 is a leucine residue;
  • Y14 is an arginine residue; and
  • Y15 is a lysine residue.
  • In a further specific embodiment, Y16 is a tryptophan, methionine or an alanine residue. Specifically, Y16 is a tryptophan residue. Alternatively, Y16 is a methionine residue.
  • In another specific embodiment,
  • Y15 is a lysine or an alanine residue;
  • Y16 is a tryptophan, methionine or an alanine residue;
  • Y17 is a methionine, norleucine, phenylalanine or an alanine residue; and
  • Y18 is a phenylalanine, isoleucine or an alanine residue.
  • In another specific embodiment, one of Y15, Y16, Y17, and Y18 is an alanine residue, provided that the other three of Y15, Y16, Y17, and Y18 are not an alanine residue.
  • In another specific embodiment,
  • Y15 is a lysine residue;
  • Y16 is a tryptophan or methionine residue;
  • Y17 is a methionine, norleucine, or phenylalanine residue; and
  • Y18 is a phenylalanine or isoleucine residue.
  • In another specific embodiment,
  • Y15 is a lysine residue;
  • Y16 is a tryptophan residue;
  • Y17 is a methionine residue; and
  • Y18 is a phenylalanine residue.
  • In another specific embodiment, Y19 is an isoleucine or alanine residue. Specifically, Y19 is an isoleucine residue.
  • In a more specific embodiment, P is selected from the group consisting of SEQ ID NOS: 24-70 as listed in Table 3 below:
  • TABLE 3
    SEQ
    i loop CRF1 Sequence ID NO:
    i2       HTAIVLTYSTDRLRKWMFI 24
    i2       HTAIVLTYSTDRLRKWM 25
    i2         AIVLTYSTDRLRKWMFI  26
    i2          GSGTYSTDRLRKWMFI 27
    i2       HTAIVLTYSTDRLRK 28
    i2          GSGTYSTDRLRKWMF 29
    i2           VLTYSTDRLRKWMFI 30
    i2       HTAIVLTYSTDRLR 31
    i2            LTYSTDRLRKWMFI 32
    i2             TYSTDRLRKWMFI 33
    i2       HTAIVLTYSTDR 34
    i2            GSGTYDRLRKWM 35
    i2          GSGTYSTDRLRK 36
    i2              GSTDRLRKWMFI 37
    i2              GSTDRLRKWMFA 38
    i2              GSTDRLRKWMAI 39
    i2              GSTDRLRKWAFI 40
    i2              GSTDRLRKAMFI 41
    i2              GSTDRLRKAXFI 42
    i2              YSTDRLRKWMFI 43
    i2              GSTDRLRKMFI 44
    i2               GSGRLRKWMFI 45
    i2               STDRLRKWMFI 46
    i2              YSTDRLRKWMF 47
    i2               STDRLRKWMF 48
    i2                ADRLRKWMFI 49
    i2                GSRLRKWMFI 50
    i2                TDRLRKWMFI 51
    i2                TDRLRKWXFI 52
    i2                TARLRKWMFI 53
    i2                TDALRKWMFI 54
    i2                TDRARKWMFI 55
    i2                TDRLAKWMFI 56
    i2                TDRLRAWMFI 57
    i2                TDRLRKAMFI 58
    i2                TDRLRKWAFI 59
    i2                TDRLRKWMAI 60
    i2                TDRLRKWMFA 61
    i2             TYSTDRLRK 62
    i2               STDRLRKMF 63
    i2               GSGRLRKWM 64
    i2                 GRLRKWMFI 65
    i2                 DRLRKWMFI 66
    i2                 DRLRKWMAI 67
    i2              GSTDRLRK 68
    i2                  RLRKWMFI 69
    i2                  RLRKWMAI 70
  • In yet another specific embodiment, P comprises at least three contiguous amino (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or 19) of the i3 intracellular loop of the CRF1 receptor.
  • In one embodiment, P is derived from the i3 loop and is represented by the following structural formula or a pharmaceutically acceptable salt thereof:
  • Z1-Z2-Z3-Z4-Z5-Z6-Z7-Z8-Z9-Z10-Z11-Z12-Z13-Z14-Z15-Z16-
    Z17-Z18-Z19-Z20-Z21-Z22-Z23-Z24-Z25-Z26-Z27-Z28-Z29-Z30-R1,
  • wherein:
  • Z1 is absent or a phenylalanine residue;
  • Z2 is absent or an asparagine residue;
  • Z3 is absent or an isoleucine residue;
  • Z4 is absent or a valine residue;
  • Z5 is absent or an arginine residue;
  • Z6 is absent or an isoleucine residue;
  • Z7 is absent or a leucine residue;
  • Z8 is absent or a methionine residue;
  • Z9 is absent or a threonine residue;
  • Z10 is absent or a lysine residue;
  • Z11 is absent or a leucine residue;
  • Z12 is absent or an arginine residue;
  • Z13 is absent or an alanine residue;
  • Z14 is absent or a serine residue;
  • Z15 is absent or a threonine residue;
  • Z16 is absent or a threonine residue;
  • Z17 is absent or a serine residue;
  • Z18 is absent or a glutamic acid residue;
  • Z19 is absent or a threonine residue;
  • Z20 is absent or an isoleucine residue;
  • Z21 is absent or a glutamine residue;
  • Z22 is absent or a tyrosine residue;
  • Z23 is absent or an arginine residue;
  • Z24 is absent or a lysine residue;
  • Z25 is absent or an alanine residue;
  • Z26 is absent or a valine residue;
  • Z27 is absent or a lysine residue;
  • Z28 is absent or an alanine or serine residue;
  • Z29 is absent or a threonine residue; and
  • Z30 is absent or a leucine residue;
  • provided that at least five of Z1-Z30 are present;
  • R1 is OR2 or N(R2)2;
  • each R2 is independently hydrogen or (C1-C10)alkyl; and
  • from 0 to 5 amino acid residues are present in the D configuration.
  • It is understood that when P is described as Z1-Z30 it is bonded to L as written. For example, Z1 is bonded to L. If Z1 is absent, then Z2 is bonded to L.
  • In a specific embodiment, at least three of Z5, Z6, Z7, Z8, and Z9 are present.
  • In a further specific embodiment,
  • Z5 is an arginine residue;
  • Z6 is an isoleucine residue; and
  • Z7 is a leucine residue.
  • In another further specific embodiment,
  • Z7 is a leucine residue;
  • Z8 is a methionine residue; and
  • Z9 is a threonine residue.
  • In a another specific embodiment,
  • Z5 is an arginine residue;
  • Z6 is an isoleucine residue;
  • Z7 is a leucine residue; and
  • Z8 is a methionine residue.
  • In a another specific embodiment,
  • Z6 is an isoleucine residue;
  • Z7 is a leucine residue;
  • Z8 is absent or a methionine residue; and
  • Z9 is a threonine residue.
  • In a another specific embodiment,
  • Z5 is an arginine residue;
  • Z6 is an isoleucine residue;
  • Z7 is a leucine residue;
  • Z8 is a methionine residue; and
  • Z9 is a threonine residue.
  • In yet another specific embodiment, at least three of Z10, Z11, Z12, and Z13 are present.
  • In a another specific embodiment,
  • Z10 is a lysine residue;
  • Z11 is a leucine residue; and
  • Z12 is an arginine residue.
  • In a another specific embodiment,
  • Z11 is a leucine residue;
  • Z12 is an arginine residue; and
  • Z13 is an alanine residue.
  • In yet another specific embodiment,
  • Z10 is a lysine residue;
  • Z11 is a leucine residue;
  • Z12 is an arginine residue; and
  • Z13 is an alanine residue.
  • In another specific embodiment, at least three of Z14, Z15, Z16, and Z17 are present.
  • In another specific embodiment,
  • Z14 is a serine residue;
  • Z15 is a threonine residue; and
  • Z16 is a threonine residue.
  • In another specific embodiment,
  • Z15 is a threonine residue;
  • Z16 is a threonine residue; and
  • Z17 is a serine residue;
  • In another specific embodiment,
  • Z14 is a serine residue;
  • Z15 is a threonine residue;
  • Z16 is a threonine residue; and
  • Z17 is a serine residue;
  • In another specific embodiment, at least three of Z19, Z20, Z21, Z22, and Z23 are present.
  • In another specific embodiment,
  • Z19 is a threonine residue;
  • Z20 is an isoleucine residue; and
  • Z21 is a glutamine residue.
  • In another specific embodiment,
  • Z21 is a glutamine residue;
  • Z22 is a tyrosine residue; and
  • Z23 is an arginine residue.
  • In another specific embodiment,
  • Z19 is a threonine residue;
  • Z20 is an isoleucine residue;
  • Z21 is a glutamine residue; and
  • Z22 is a tyrosine residue.
  • In another specific embodiment,
  • Z20 is an isoleucine residue;
  • Z21 is a glutamine residue;
  • Z22 is a tyrosine residue; and
  • Z23 is an arginine residue.
  • In another specific embodiment,
  • Z19 is a threonine residue;
  • Z20 is an isoleucine residue;
  • Z21 is a glutamine residue;
  • Z22 is a tyrosine residue; and
  • Z23 is an arginine residue.
  • In another specific embodiment, at least three of Z24, Z25, Z26, Z27, and Z28 are present.
  • In another specific embodiment,
  • Z24 is a lysine residue;
  • Z25 is an alanine residue; and
  • Z26 is a valine residue.
  • In another specific embodiment,
  • Z26 is a valine residue;
  • Z27 is a lysine residue; and
  • Z28 is an alanine or serine residue.
  • In another specific embodiment,
  • Z24 is a lysine residue;
  • Z25 is an alanine residue;
  • Z26 is a valine residue; and
  • Z27 is a lysine residue.
  • In another specific embodiment,
  • Z25 is an alanine residue;
  • Z26 is a valine residue;
  • Z27 is a lysine residue; and
  • Z28 is an alanine or serine residue.
  • In another specific embodiment,
  • Z24 is a lysine residue;
  • Z25 is an alanine residue;
  • Z26 is a valine residue;
  • Z27 is a lysine residue; and
  • Z28 is an alanine or serine residue. Specifically, Z28 is an alanine residue.
  • In a more specific embodiment, P is selected from the group consisting of SEQ ID NOS: 71-98 as listed in Table 4 below:
  • TABLE 4
    SEQ
    i loop Sequence ID NO:
    i3    FNIVRILMTKLRASTTSETIQYRKAVKA 71
    i3      IVRILMTKLRASTTSETIQYRKAVKA 72
    i3    FNIVRILMTKLRASTTSETIQYRKAV 73
    i3        RILMTKLRASTTSETIQYRKAVKA 74
    i3    FNIVRILMTKLRASTTSETIQYRK 75
    i3    FNIVRILMTKLRASTTSETIQYR 76
    i3          LMTKLRASTTSETIQYRKAVKA 77
    i3    FNIVRILMTKLRASTTSETIQY 78
    i3            TKLRASTTSETIQYRKAVKA 79
    i3          LMTKLRASTTSETIQYRKAV 80
    i3              LRASTTSETIQYRKAVKA 81
    i3          LMTKLRASTTSETIQYRK 82
    i3          LMTKLRASTTSETIQYR 83
    i3                ASTTSETIQYRKAVKA 84
    i3                ASTTSETIQYRKAVKS 85
    i3          LMTKLRASTTSETIQ 86
    i3                    SETIQYRKAVKATL 87
    i3          LMTKLRASTTSETI 88
    i3        RILMTKLRASTTS 89
    i3                    SETIQYRKAVKA 90
    i3            TKLRASTTSETI 91
    i3                ASTTSETIQYR 92
    i3            TKLRASTTSET 93
    i3             KLRASTTSETI 94
    i3                      TIQYRKAVKA 95
    i3             KLRASTTSET 96
    i3        RILMTKLRA 97
    i3             KLRASTTSE 98
  • In further specific embodiment, P comprises at least three contiguous amino (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or 19) of the i4 intracellular domain of the CRF1 receptor.
  • In one embodiment, P derived from the i4 domain and is represented by the following structural formula or a pharmaceutically acceptable salt thereof
  • M1-M2-M3-M4-M5-M6-M7-M8-M9-M10-M11-M12-M13-M14-M15-M16-
    M17-M18-M19-M20-M21-M22-M23-M24-M25-M26-M27-M28-M29-M30-
    M31-M32-M33-M34-M35-M36-M37-M38-M39-M40-M41-M42-M43-M44-
    M45-M46-M47-R1,
  • wherein:
  • M1 is absent or a glutamic acid residue;
  • M2 is absent or a valine residue;
  • M3 is absent or an arginine residue;
  • M4 is absent or a serine residue;
  • M5 is absent or an alanine residue;
  • M6 is absent or an isoleucine residue;
  • M7 is absent or an arginine residue;
  • M8 is absent or a lysine residue;
  • M9 is absent or an arginine residue;
  • M10 is absent or a tryptophan residue;
  • M11 is absent or a histidine residue;
  • M12 is absent or an arginine residue;
  • M13 is absent or a tryptophan residue;
  • M14 is absent or a glutamine residue;
  • M15 is absent or an aspartic acid residue;
  • M16 is absent or a lysine residue;
  • M17 is absent or a histidine or glycine residue;
  • M18 is absent or a serine residue;
  • M19 is absent or an isoleucine residue;
  • M20 is absent or an arginine residue;
  • M21 is absent or an alanine residue;
  • M22 is absent or an arginine residue;
  • M23 is absent or a valine residue;
  • M24 is absent or an alanine residue;
  • M25 is absent or an arginine residue;
  • M26 is absent or an alanine residue;
  • M27 is absent or a methionine residue;
  • M28 is absent or a serine residue;
  • M29 is absent or an isoleucine residue;
  • M30 is absent or a proline residue;
  • M31 is absent or a threonine residue;
  • M32 is absent or a serine residue;
  • M33 is absent or a proline residue;
  • M34 is absent or a threonine residue;
  • M35 is absent or an arginine residue;
  • M36 is absent or a valine residue;
  • M37 is absent or a serine residue;
  • M38 is absent or a phenylalanine residue;
  • M39 is absent or a histidine residue;
  • M40 is absent or a serine residue;
  • M41 is absent or an isoleucine residue;
  • M42 is absent or a lysine residue;
  • M43 is absent or a glutamine residue;
  • M44 is absent or a serine residue;
  • M45 is absent or a threonine residue;
  • M46 is absent or an alanine residue; and
  • M47 is absent or a valine residue; provided that at least five of M1-M47 are present;
  • R1 is OR2 or N(R2)2;
  • each R2 is independently hydrogen or (C1-C10)alkyl; and
  • from 0 to 5 amino acid residues are present in the D configuration.
  • It is understood that when P is described as M1-M47 it is bonded to L as written. For example, M1 is bonded to L. If M1 is absent, then M2 is bonded to L.
  • In a specific embodiment, at least three of M2-M7 are present.
  • In another specific embodiment,
  • M2 is a valine residue;
  • M3 is an arginine residue; and
  • M4 is a serine residue.
  • In another specific embodiment,
  • M5 is an alanine residue;
  • M6 is an isoleucine residue; and
  • M7 is an arginine residue.
  • In another specific embodiment,
  • M2 is a valine residue;
  • M3 is an arginine residue;
  • M4 is a serine residue; and
  • M5 is an alanine residue.
  • In another specific embodiment,
  • M4 is a serine residue;
  • M5 is an alanine residue;
  • M6 is an isoleucine residue; and
  • M7 is an arginine residue.
  • In another specific embodiment,
  • M2 is a valine residue;
  • M3 is an arginine residue;
  • M4 is a serine residue;
  • M5 is an alanine residue;
  • M6 is an isoleucine residue.
  • In another specific embodiment,
  • M3 is an arginine residue;
  • M4 is a serine residue;
  • M5 is an alanine residue;
  • M6 is an isoleucine residue; and
  • M7 is an arginine residue.
  • In another specific embodiment,
  • M2 is a valine residue;
  • M3 is an arginine residue;
  • M4 is a serine residue;
  • M5 is an alanine residue;
  • M6 is an isoleucine residue; and
  • M7 is an arginine residue.
  • In another specific embodiment, at least three of M9-M13 are present.
  • In another specific embodiment,
  • M9 is an arginine residue;
  • M10 is a tryptophan residue;
  • M11 is a histidine residue;
  • M12 is an arginine residue.
  • In another specific embodiment,
  • M10 is a tryptophan residue;
  • M11 is a histidine residue;
  • M12 is an arginine residue; and
  • M13 is a tryptophan residue.
  • In another specific embodiment,
  • M9 is an arginine residue;
  • M10 is a tryptophan residue;
  • M11 is a histidine residue;
  • M12 is an arginine residue; and
  • M13 is a tryptophan residue.
  • In another specific embodiment, at least three of M18-M22 are present.
  • In another specific embodiment,
  • M18 is a serine residue;
  • M19 is an isoleucine residue;
  • M20 is an arginine residue; and
  • M21 is an alanine residue.
  • In another specific embodiment,
  • M19 is an isoleucine residue;
  • M20 is an arginine residue;
  • M21 is an alanine residue; and
  • M22 is an arginine residue.
  • In another specific embodiment,
  • M18 is a serine residue;
  • M19 is an isoleucine residue;
  • M20 is an arginine residue;
  • M21 is an alanine residue; and
  • M22 is an arginine residue.
  • In another specific embodiment, at least three of M23-M27 are present.
  • In another specific embodiment,
  • M23 is a valine residue;
  • M24 is an alanine residue;
  • M25 is an arginine residue; and
  • M26 is an alanine residue.
  • In another specific embodiment,
  • M24 is an alanine residue;
  • M25 is an arginine residue;
  • M26 is an alanine residue;
  • M27 is a methionine residue.
  • In another specific embodiment,
  • M23 is a valine residue;
  • M24 is an alanine residue;
  • M25 is an arginine residue;
  • M26 is an alanine residue;
  • M27 is a methionine residue.
  • In another specific embodiment, at least three of M25-M29 are present.
  • In another specific embodiment,
  • M25 is an arginine residue;
  • M26 is an alanine residue;
  • M27 is a methionine residue; and
  • M28 is a serine residue.
  • In another specific embodiment,
  • M26 is an alanine residue;
  • M27 is a methionine residue;
  • M28 is a serine residue; and
  • M29 is an isoleucine residue.
  • In another specific embodiment,
  • M25 is an arginine residue;
  • M26 is an alanine residue;
  • M27 is a methionine residue;
  • M28 is a serine residue; and
  • M29 is an isoleucine residue.
  • In another specific embodiment, at least three of M32-M36 are present.
  • In another specific embodiment,
  • M32 is a serine residue;
  • M33 is a proline residue;
  • M34 is a threonine residue; and
  • M35 is an arginine residue.
  • In another specific embodiment,
  • M33 is a proline residue;
  • M34 is a threonine residue;
  • M35 is an arginine residue; and
  • M36 is a valine residue.
  • In another specific embodiment,
  • M32 is a serine residue;
  • M33 is a proline residue;
  • M34 is a threonine residue;
  • M35 is an arginine residue; and
  • M36 is a valine residue.
  • In a more specific embodiment, P is selected from the group consisting of SEQ ID NOS: 99-106 as listed in Table 5 below:
  • TABLE 5
    SEQ
    i loop CRF1 Sequence ID:
    i4 EVRSAIRKRWHRWQDKHSIRARVARAMSIPTSPTRVSFHSIKQSTAV 99
    i4                         RAMSIPTSPTRVSFHSIKQSTAV 100
    i4                 HSIRARVARAMSIPTSPTRV 101
    i4            RWQDKHSIRARVARAMSI 102
    i4 EVRSAIRKRWHRWQD 103
    i4         RWHRWQDKHSIRAR 104
    i4  VRSAIRKRWHRW 105
    i4                 GSIRARVARAM 106
  • It is understood that the sequences presented in Tables 2-5 can be optionally functionalized at the C-terminus. Functionalized at the C-terminus means that the acid moiety present at the C-terminus is replaced by some other functional group. Suitable functional groups include —C(O)N(R2)2, —C(O)OR3, or C(O)NHC(O)OR2, where R2 is hydrogen or a (C1-C10) alkyl group and R3 is a (C1-C10) alkyl group.
  • It is understood that as long as P comprises the indicated number of contiguous amino acids residues from the CRF1 intracellular loop (i1, i2 or i3) or domain (i4) from which it is derived, the remainder of the peptide, if present, can be selected from:
  • (a) any natural amino acid residue, unnatural amino acid residue or a combination thereof;
  • (b) a peptide sequence comprising natural amino acid residues, non-natural amino acid residues and combinations thereof;
  • (c) a peptide sequence according to (b) comprising one or more peptide backbone modifications;
  • (d) a peptide sequence according to (c) comprising one or more retro-inverso peptide linkages;
  • (e) a peptide sequence according to (c) wherein one or more peptide bonds are replaced by
  • Figure US20110301087A1-20111208-C00001
  • or a combination thereof;
  • (f) a peptide sequence according to (c) comprising one or more depsipeptide linkages, wherein the amide linkage is replaced with an ester linkage; and
  • (g) a peptide sequence according to (c) comprising one or more conformational restrictions; and
  • (h) a peptide sequence according to (c) comprising one or more of (d)-(g).
  • Furthermore, it is understood that even within the indicated number of contiguous amino acid residues derived from the GPCR intracellular loop (i1, i2 or i3) or domain (i4), there can be: peptide backbone modifications such as, but not limited to, those described in (e) above; retro-inverso peptide linkages; despsipeptide linkages; conformational restrictions; or a combination thereof.
  • It is noted that P of Formula I can be optionally functionalized at the C-terminus. Functionalized at the C-terminus means that the acid moiety present at the C-terminus is replaced by some other functional group. Suitable functional groups include —C(O)N(R2)2, —C(O)OR3, or C(O)NHC(O)OR2, where R2 is hydrogen or a (C1-C10) alkyl group and R3 is a (C1-C10) alkyl group. Functionalization of the C-terminus can result from the methods used to prepare.
  • Peptidomimetic as used herein refers to a compound comprising non-peptidic structural elements in place of a peptide sequence.
  • As used herein, the term “amino acid” includes both a naturally occurring amino acid and a non-natural amino acid.
  • As used herein, the term “naturally occurring amino acid” means a compound represented by the formula NH2—CHR—COOH, wherein R is the side chain of a naturally occurring amino acids such as lysine, arginine, serine, tyrosine etc. as shown in the Table below.
  • Table of Common Naturally Occurring Amino Acids
    Amino acid Three letter code One letter code
    Non-polar; alanine Ala A
    neutral at isoleucine Ile I
    pH 7.4 leucine Leu L
    methionine Met M
    phenylalanine Phe F
    proline Pro P
    tryptophan Trp W
    valine Val V
    Polar, asparagine Asn N
    uncharged cysteine Cys C
    at pH 7.0 glycine Gly G
    glutamine Gln Q
    serine Ser S
    threonine Thr T
    tyrosine Tyr Y
    Polar; glutamic acid Glu E
    charged at arginine Arg R
    pH 7 aspartic acid Asp D
    histidine His H
    lysine Lys K
  • “Non-natural amino acid” means an amino acid for which there is no nucleic acid codon. Examples of non-natural amino acids include, for example, the D-isomers of the natural α-amino acids such as D-proline (D-P, D-Pro) as indicated above; natural α-amino acids with non-natural side chains (e.g.,
  • Figure US20110301087A1-20111208-C00002
  • related to phenylalanine); Aib (aminobutyric acid), bAib (3-aminoisobutyric acid), Nva (norvaline), β-Ala, Aad (2-aminoadipic acid), bAad (3-aminoadipic acid), Abu (2-aminobutyric acid), Gaba (γ-aminobutyric acid), Acp (6-aminocaproic acid), Dbu (2,4-diaminobutryic acid), α-aminopimelic acid, TMSA (trimethylsilyl-Ala), aIle (allo-isoleucine), Nle (norleucine), tert-Leu, Cit (citrulline), Orn (ornithine, O), Dpm (2,2′-diaminopimelic acid), Dpr (2,3-diaminopropionic acid), α or .β-Nal, Cha (cyclohexyl-Ala), hydroxyproline, Sar (sarcosine), and the like.
  • Unnatural amino acids also include cyclic amino acids; and amino acid analogs, for example, Nα-alkylated amino acids such as MeGly (Nα-methylglycine), EtGly (Nα-ethylglycine) and EtAsn (Nα-ethylasparagine); and amino acids in which the α-carbon bears two side-chain substituents. As with the natural amino acids, the residues of the unnatural amino acids are what are left behind when the unnatural amino acid becomes part of a peptide sequence as described herein.
  • Amino acid residues are amino acid structures as described above that lack a hydrogen atom of the amino group or the hydroxyl moiety of the carboxyl group or both resulting in the units of a peptide chain being amino-acid residues.
  • The D-isomers of the natural amino acids are designated herein with a lower case letter of the corresponding naturally occurring amino acid. For example, d-proline is designated “p” rather than “P” as is used for naturally occurring proline.
  • Tethers (T)
  • T of Formula I is a lipohilic tether moiety which imparts lipophilicity to the CRF1 receptor compounds of the invention. The lipophilicity which T imparts, can promote penetration of the CRF1 receptor compounds into the cell membrane and tethering of the CRF1 receptor compounds to the cell membrane. As such, the lipophilicity imparted by T can facilitate interaction between the CRF1 receptor compounds of the invention and the cognate receptor.
  • The relative lipophilicity of compounds suitable for use as the lipophilic tether moiety of Formula I can be quantified by measuring the amount of the compound that partitions into an organic solvent layer (membrane-like) vs. an aqueous solvent layer (analogous to the extracellular or cytoplasmic environment). The partition coefficient in a mixed solvent composition, such as octanol/water or octanol/PBS, is the ratio of compound found at equilibrium in the octanol vs. the aqueous solvent (Partition coeff P=[compound]octanol/[compound]aqueous). Frequently, the partition coefficient is expressed in logarithmic form, as the log P. Compounds with greater lipophilicity have a more positive log P than more hydrophilic compounds and tend to interact more strongly with membrane bilayers.
  • Computational programs are also available for calculating the partition coefficient for compounds suitable for use as the lipophilic tether moiety (T). In situations where the chemical structure is being varied in a systematic manner, for example by adding additional methylene units (—CH2—) onto to an existing alkyl group, the trend in log P can be calculated using, for example, ChemDraw (CambridgeSoft, Inc).
  • In one embodiment, T is an optionally substituted (C6-C30)alkyl, (C6-C30)alkenyl, (C6-C30)alkynyl wherein 0-3 carbon atoms are replaced with oxygen, sulfur, nitrogen or a combination thereof.
  • In a specific embodiment, the (C6-C30)alkyl, (C6-C30)alkenyl, (C6-C30)alkynyl are substituted at one or more substitutable carbon atoms with halogen, —CN, —OH, —NH2, NO2, —NH(C1-C6)alkyl, —N((C1-C6)alkyl)2, (C1-C6)alkyl, (C1-C6)haloalkyl, (C1-C6)alkoxy, (C1-C6)haloalkoxy, aryloxy, (C1-C6)alkoxycarbonyl, —CONH2, —OCONH2, —NHCONH2, —N(C1-C6)alkylCONH2, —N(C1-C6)alkylCONH(C1-C6)alkyl, —NHCONH(C1-C6)alkyl, —NHCON((C1-C6)alkyl)2, —N(C1-C6)alkylCON((C1-C6)alkyl)2, —NHC(S)NH2, —N(C1-C6)alkylC(S)NH2, —N(C1-C6)alkylC(S)NH(C1-C6)alkyl, —NHC(S)NH(C1-C6)alkyl, —NHC(S)N((C1-C6)alkyl)2, —N(C1-C6)alkylC(S)N((C1-C6)alkyl)2, —CONH(C1-C6)alkyl, —OCONH(C1-C6)alkyl —CON((C1-C6)alkyl)2, —C(S)(C1-C6)alkyl, —S(O)p(C1-C6)alkyl, —S(O)pNH2, —S(O)pNH(C1-C6)alkyl, —S(O)pN((C1-C6)alkyl)2, —CO(C1-C6)alkyl, —OCO(C1-C6)alkyl, —C(O)O(C1-C6)alkyl, —OC(O)O(C1-C6)alkyl, —C(O)H or —CO2H; and p is 1 or 2.
  • In a specific embodiment, T is selected from the group consisting of: CH3(CH2)9OPh-, CH3(CH2)6C═C(CH2)6, CH3(CH2)11O(CH2)3, CH3(CH2)9O(CH2)2 and CH3(CH2)13.
  • In a specific embodiment, T is selected from the group consisting of: CH3(CH2)16, CH3(CH2)15, CH3(CH2)14, CH3(CH2)13, CH3(CH2)12, CH3(CH2)11, CH3(CH2)10, CH3(CH2)9, CH3(CH2)8, CH3(CH2)9OPh-, CH3(CH2)6C═C(CH2)6, CH3(CH2)11O(CH2)3, and CH3(CH2)9O(CH2)2 and CH3(CH2)13.
  • It is understood that the lipophilic moiety (T) of Formula I can be derived from precursor liphophilic compounds (e.g., fatty acids and bile acids). As used herein, “derived from” with regard to T, means that T is derived from a precursor lipophilic compound and that reaction of the precursor lipophilic compound in preparing the CRF1 receptor compounds of Formula I, results in a lipophilic tether moiety represented by T in Formula I that is structurally modified in comparison to the precursor lipophilic compound.
  • For example, the lipophilic tether moiety, T of Formula I, can be derived from a fatty acid or a bile acid. It is understood that in accordance with Formula I, when T is derived from a fatty acid (i.e., a fatty acid derivative) it is attached to L-P at the carbon atom alpha to the carbonyl carbon of the acid functional group in the fatty acid from which it is derived. For example, when T is derived from palmitic acid,
  • Figure US20110301087A1-20111208-C00003
  • T of Formula I has the following structure:
  • Figure US20110301087A1-20111208-C00004
  • Similarly, when T is derived from stearic acid,
  • Figure US20110301087A1-20111208-C00005
  • T of Formula I has the following structure:
  • Figure US20110301087A1-20111208-C00006
  • Similarly, when T is derived from 3-(dodecyloxy)propanoic acid,
  • Figure US20110301087A1-20111208-C00007
  • T of Formula I has the following structure:
  • Figure US20110301087A1-20111208-C00008
  • Similarly, when T is derived from 4-(undecyloxy)butanoic acid,
  • Figure US20110301087A1-20111208-C00009
  • T of Formula I has the following structure:
  • Figure US20110301087A1-20111208-C00010
  • Similarly, when T is derived from elaidic acid,
  • Figure US20110301087A1-20111208-C00011
  • T of Formula I has the following structure:
  • Figure US20110301087A1-20111208-C00012
  • Similarly, when T is derived from oleic acid,
  • Figure US20110301087A1-20111208-C00013
  • T of Formula I has the following structure:
  • Figure US20110301087A1-20111208-C00014
  • Similarly, when T is derived from 16-hydroxypalmitic acid,
  • Figure US20110301087A1-20111208-C00015
  • T of Formula I has the following structure:
  • Figure US20110301087A1-20111208-C00016
  • Similarly, when T is derived from 2-aminooctadecanoic acid
  • Figure US20110301087A1-20111208-C00017
  • T of Formula I has the following structure:
  • Figure US20110301087A1-20111208-C00018
  • Similarly, when T is derived from 2-amino-4-(dodecyloxy)butanoic acid
  • Figure US20110301087A1-20111208-C00019
  • T of Formula I has the following structure:
  • Figure US20110301087A1-20111208-C00020
  • In a further embodiment, T is derived from a fatty acid. In a specific embodiment, T is derived from a fatty acid selected from the group consisting of: butyric acid, caproic acid, caprylic acid, capric acid, lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, and lignoceric acid.
  • In another specific embodiment, T is derived from a fatty acid selected from the group consisting of: myristoleic acid, palmitoleic acid, oleic acid, linoleic acid, α-linolenic acid, arachidonic acid, eicosapentaenoic acid, erucic acid, docosahexaenoic acid
  • In another embodiment, T of Formula I can be derived from a bile acid. Similar to the embodiment where T is a fatty acid derivative, it is understood that in accordance with Formula I, when T is derived from a bile acid (i.e., a bile acid derivative) it is attached to L-P at the carbon atom alpha to the carbonyl carbon of the acid functional group in the bile acid from which it is derived. For example, when T is derived from lithocholic acid,
  • Figure US20110301087A1-20111208-C00021
  • T of Formula I has the following structure:
  • Figure US20110301087A1-20111208-C00022
  • In a further embodiment, T is derived from a bile acid. In a specific embodiment, T is derived from a bile acid selected from the group consisting of: lithocholic acid, chenodeoxycholic acid, deoxycholic acid, cholanic acid, cholic acid, ursocholic acid, ursodeoxycholic acid, isoursodeoxycholic acid, lagodeoxycholic acid, dehydrocholic acid, hyocholic acid, hyodeoxycholic acid and the like.
  • For example, T is selected from:
  • Figure US20110301087A1-20111208-C00023
  • In another further embodiment, T is derived from a bile acid described above that has been modified at other than the acid functional group. For example, T can be derived from any of the bile acids described above, where the hydroxy position has been modified to form an ester or a halo ester. For example, T can be:
  • Figure US20110301087A1-20111208-C00024
  • Other lipophilic moieties suitable for use as the lipophilic membrane tether, T, of Formula I, include but are not limited to steroids. Suitable steroids include, but are not limited to, sterols; progestagens; glucocorticoids; mineralcorticoids; androgens; and estrogens. Generally any steroid capable of attachment or which can be modified for incorporation into Formula I can be used. It is understood that the lipophilic membrane tether, T, may be slightly modified from the precursor lipophilic compound as a result of incorporation into Formula I.
  • Suitable sterols for use in the invention at T, include but are not limited to: cholestanol, coprostanol, cholesterol, epicholesterol, ergosterol, ergocalciferol, and the like. Preferred sterols are those that provide a balance of lipophilicity with water solubility.
  • Suitable progestagens include, but are not limited to progesterone. Suitable glucocorticoids include, but are not limited to cortisol. Suitable mineralcorticoids include, but are not limited to aldosterone. Suitable androgens include, but are not limited to testosterone and androstenedione. Suitable estrogens include, but are not limited to estrone and estradiol.
  • In another specific embodiment, T can be derived from 2-tetradecanamideooctadecanoid acid. Similar to the embodiment where T is a fatty acid derivative, it is understood that in accordance with Formula I, when T is derived from 2-tetradecanamideooctadecanoid acid it is attached to L-P at the carbon atom alpha to the carbonyl carbon of the acid functional group in the bile acid from which it is derived. For example, when T is derived from 2-tetradecanamideooctadecanoid acid, the tether is:
  • Figure US20110301087A1-20111208-C00025
  • In another embodiment, T of Formula I can be derived from 2-(5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamido)octadecanoic acid. For example, when T is derived from 2-(5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamido)octadecanoic acid, the tether is:
  • Figure US20110301087A1-20111208-C00026
  • In yet another embodiment, T of Formula I can be:
  • Figure US20110301087A1-20111208-C00027
  • It is understood, that the compounds can contain one of more tether moieties. In certain aspects, the tether moieties are the same. In other embodiments, the tether moieties are different.
  • Compounds (T-L-P)
  • In a first aspect, the GPCR Compound of the invention is represented by Formula I:

  • T-L-P,
  • or pharmaceutically acceptable salts thereof, wherein:
      • P is a peptide comprising at least three contiguous amino-acid residues of an intracellular i1, i2, i3 loop or an intracellular i4 domain of the CRF1 receptor;
      • L is a linking moiety represented by C(O) and bonded to P at an N terminal nitrogen of an N-terminal amino-acid residue;
      • and T is a lipophilic tether moiety bonded to L, wherein the C-terminal amino acid residue of P is optionally functionalized.
  • In a second aspect, P comprises at least six contiguous amino acid residues.
  • In a third aspect, P comprises at least 3 contiguous amino acids of the i1 loop.
  • In a specific embodiment of the third aspect, P is derived from the i1 loop and is represented by the following structural formula or pharmaceutically acceptable salts thereof
  • X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11-X12-X13-X14-X15-X16-
    X17-X18-X19-R1
  • wherein:
      • X1 is absent or a valine residue;
      • X2 is absent or a leucine or glycine residue;
      • X3 is absent or a phenylalanine or serine residue;
      • X4 is absent or a leucine or glycine residue;
      • X5 is absent or an arginine residue;
      • X6 is absent or a leucine or isoleucine residue;
      • X7 is absent or an arginine residue;
      • X8 is absent or a serine residue;
      • X9 is absent or an isoleucine residue;
      • X10 is absent or an arginine residue;
      • X11 is absent or a cysteine or serine residue;
      • X12 is absent or a leucine residue;
      • X13 is absent or an arginine residue;
      • X14 is absent or an asparagine residue;
      • X15 is absent or an isoleucine residue;
      • X16 is absent or an isoleucine residue;
      • X17 is absent or a histidine residue;
      • X18 is absent or tryptophan residue;
      • X19 is absent or an asparagine residue; provided that at least five of X1-X19 are present;
      • R1 is OR2 or N(R2)2;
      • each R2 is independently hydrogen or (C1-C10)alkyl; and
      • from 0 to 5 amino acid residues are present in the D configuration.
  • In a specific embodiment, at least four of X7, X8, X9, X10, X11, X12, X13 and X14 are present.
  • In a further specific embodiment,
  • X7 is an arginine residue;
  • X8 is a serine residue;
  • X9 is an isoleucine residue; and
  • X10 is an arginine residue.
  • In another further specific embodiment,
  • X11 is a cysteine or serine residue;
  • X12 is a leucine residue;
  • X13 is an arginine residue; and
  • X14 is an asparagine residue.
  • In another further specific embodiment, X11 is a cysteine residue. Alternatively, wherein X11 is a serine residue.
  • In another specific embodiment,
  • X14 is an asparagine residue;
  • X15 is an isoleucine residue;
  • X16 is an isoleucine residue; and
  • X17 is a histidine residue.
  • In a specific embodiment of the third aspect, the i1 loop of the CRF1 receptor from which P is derived has the following sequence: VLFLRLRSIRCLRNIIHWN (SEQ ID NO: 1).
  • In another embodiment of the third aspect, P is a sequence selected from:
  • VLFLRLRSIRCLRNIIHWN; (SEQ ID NO: 1)
    VLFLRLRSIRCLRNIIHW; (SEQ ID NO: 2)
    GSGRLRSIRSLRNIIHWN; (SEQ ID NO: 3)
    VLFLRLRSIRCLRNIIH; (SEQ ID NO: 4)
    FLRLRSIRCLRNIIHWN; (SEQ ID NO: 5)
    GSGRLRSIRSLRNIIH; (SEQ ID NO: 6)
    LFIRIRSIRSLRNIIH; (SEQ ID NO: 7)
    VLFLRLRSIRCLRNII; (SEQ ID NO: 8)
    RLRSIRCLRNIIHWN; (SEQ ID NO: 9)
    RLRSIRSLRNIIHWN; (SEQ ID NO: 10)
    VLFLRLRSIRCLRNI; (SEQ ID NO: 11)
    VLFLRLRSIRCLRN; (SEQ ID NO: 12)
    RLRSIRSLRNIIH; (SEQ ID NO: 13)
    RSIRSLRNIIHWN; (SEQ ID NO: 14)
    VLFLRLRSIRCLR; (SEQ ID NO: 15)
    RSIRCLRNIIHW; (SEQ ID NO: 16)
    RSIRSLRNIIH; (SEQ ID NO: 17)
    RSIRCLRNIIH; (SEQ ID NO: 18)
    RLRSIRSLRN; (SEQ ID NO: 19)
    RSLRNIIHWN; (SEQ ID NO: 20)
    RLRSIRSLR; (SEQ ID NO: 21)
    RSLRNIIH; (SEQ ID NO: 22)
    and
    RSIRSLRN. (SEQ ID NO: 23)
  • In a fourth aspect, P comprises at least 3 contiguous amino acids of the i2 loop.
  • In a specific embodiment of the fourth aspect, P derived from the i2 loop and is represented by the following structural formula or a pharmaceutically acceptable salt thereof
  • Y1-Y2-Y3-Y4-Y5-Y6-Y7-Y8-Y9-Y10-Y11-Y12-Y13-Y14-Y15-Y16-
    Y17-Y18-Y19-R1,
  • wherein:
      • Y1 is absent or a histidine residue;
      • Y2 is absent or a threonine residue;
      • Y3 is absent or an alanine residue;
      • Y4 is absent or an isoleucine or a glycine residue;
      • Y5 is absent or a valine or serine residue;
      • Y6 is absent or a leucine or glycine residue;
      • Y7 is absent or a threonine or serine residue;
      • Y8 is absent or a tyrosine or glycine residue;
      • Y9 is absent or serine, threonine or glycine residue;
      • Y10 is absent or a threonine, serine, tyrosine, glycine or alanine residue;
      • Y11 is absent or an aspartic acid, alanine, serine or glycine residue;
      • Y12 is absent or an arginine or alanine residue;
      • Y13 is absent or a leucine or an alanine residue;
      • Y14 is absent or an arginine or alanine residue;
      • Y15 is absent or a lysine or an alanine residue;
      • Y16 is absent or a tryptophan, methionine or an alanine residue;
      • Y17 is absent or a methionine, norleucine, phenylalanine or an alanine residue;
      • Y18 is absent or a phenylalanine, isoleucine or an alanine residue;
      • Y19 is absent or an isoleucine or alanine residue, provided that at least five of Y1-Y19, are present; and
      • R1 is OR2 or N(R2)2;
      • each R2 is independently hydrogen or (C1-C10)alkyl; and
      • from 0 to 5 amino acid residues are present in the D configuration.
  • It is understood that when P is described as Y1-Y19 it is bonded to L as written. For example, Y1 is bonded to L. If Y1 is absent, then Y2 is bonded to L.
  • In a specific embodiment, at least three of Y7, Y8, Y9, Y10, Y11, Y12, Y13, Y14, Y15, Y16, Y17, Y18 and Y19 are present.
  • In a further specific embodiment,
  • Y9 is serine, threonine or glycine residue;
  • Y10 is a threonine, serine, tyrosine, glycine or alanine residue; and
  • Y11 is an aspartic acid, alanine, serine or glycine residue.
  • In another further specific embodiment,
  • Y12 is an arginine or alanine residue;
  • Y13 is a leucine or an alanine residue; and
  • Y14 is an arginine or alanine residue.
  • In a further specific embodiment,
  • Y9 is serine, threonine or glycine residue;
  • Y10 is a threonine, serine, tyrosine, glycine or alanine residue;
  • Y11 is an aspartic acid, alanine, serine or glycine residue; and
  • Y12 is an arginine or alanine residue.
  • In a further specific embodiment,
  • Y11 is an aspartic acid, alanine, serine or glycine residue;
  • Y12 is an arginine or alanine residue;
  • Y13 is a leucine or an alanine residue; and
  • Y14 is an arginine or alanine residue.
  • In a further specific embodiment,
  • Y9 is serine, threonine or glycine residue;
  • Y10 is a threonine, serine, tyrosine, glycine or alanine residue;
  • Y11 is an aspartic acid, alanine, serine or glycine residue;
  • Y12 is an arginine or alanine residue; and
  • Y13 is a leucine or an alanine residue.
  • In a further specific embodiment,
  • Y10 is a threonine, serine, tyrosine, glycine or alanine residue;
  • Y11 is an aspartic acid, alanine, serine or glycine residue;
  • Y12 is an arginine or alanine residue;
  • Y13 is a leucine or an alanine residue; and
  • Y14 is an arginine or alanine residue.
  • In yet another further specific embodiment,
  • Y9 is serine, threonine or glycine residue;
  • Y10 is a threonine, serine, tyrosine, glycine or alanine residue;
  • Y11 is an aspartic acid, alanine, serine or glycine residue;
  • Y12 is an arginine or alanine residue;
  • Y13 is a leucine or an alanine residue; and
  • Y14 is an arginine or alanine residue.
  • In yet another further specific embodiment,
  • Y9 is serine residue;
  • Y10 is a threonine residue;
  • Y11 is an aspartic acid residue;
  • Y12 is an arginine residue;
  • Y13 is a leucine residue; and
  • Y14 is an arginine residue.
  • In a specific embodiment,
  • Y7 is a threonine or serine residue;
  • Y8 is a tyrosine or glycine residue;
  • Y9 is a serine, threonine or glycine residue; and
  • Y10 is a threonine, serine, tyrosine, glycine or alanine residue.
  • In another specific embodiment,
  • Y7 is a threonine residue;
  • Y8 is a tyrosine residue;
  • Y9 is a serine residue; and
  • Y10 is a threonine residue.
  • In a further specific embodiment, Y11 is an aspartic acid, alanine, serine or glycine residue. Specifically, Y11 is an aspartic acid residue.
  • In another specific embodiment,
  • Y12 is an arginine or alanine residue;
  • Y13 is or a leucine or an alanine residue;
  • Y14 is an arginine or alanine residue; and
  • Y15 is a lysine or an alanine residue.
  • In another specific embodiment, one of Y12, Y13, Y14, and Y15 is an alanine residue, provided that the other three of Y12, Y13, Y14, and Y15 are not an alanine residue.
  • In another specific embodiment,
  • Y12 is an arginine residue;
  • Y13 is a leucine residue;
  • Y14 is an arginine residue; and
  • Y15 is a lysine residue.
  • In a further specific embodiment, Y16 is a tryptophan, methionine or an alanine residue. Specifically, Y16 is a tryptophan residue. Alternatively, Y16 is a methionine residue.
  • In another specific embodiment,
  • Y15 is a lysine or an alanine residue;
  • Y16 is a tryptophan, methionine or an alanine residue;
  • Y17 is a methionine, norleucine, phenylalanine or an alanine residue; and
  • Y18 is a phenylalanine, isoleucine or an alanine residue.
  • In another specific embodiment, one of Y15, Y16, Y17, and Y18 is an alanine residue, provided that the other three of Y15, Y16, Y17, and Y18 are not an alanine residue.
  • In another specific embodiment,
  • Y15 is a lysine residue;
  • Y16 is a tryptophan or methionine residue;
  • Y17 is a methionine, norleucine, or phenylalanine residue; and
  • Y18 is a phenylalanine or isoleucine residue.
  • In another specific embodiment,
  • Y15 is a lysine residue;
  • Y16 is a tryptophan residue;
  • Y17 is a methionine residue; and
  • Y18 is a phenylalanine residue.
  • In another specific embodiment, Y19 is an isoleucine or alanine residue. Specifically, Y19 is an isoleucine residue.
  • In a specific embodiment of the fourth aspect, the i2 loop of the CRF1 receptor from which P is derived has the following sequence: HTAIVLTYSTDRLRKWMFI (SEQ ID NO: 24).
  • In another embodiment of the fourth aspect, P is a sequence selected from:
  • HTAIVLTYSTDRLRKWMFI; (SEQ ID NO: 24)
    HTAIVLTYSTDRLRKWM; (SEQ ID NO: 25)
    AIVLTYSTDRLRKWMFI; (SEQ ID NO: 26)
    GSGTYSTDRLRKWMFI; (SEQ ID NO: 27)
    HTAIVLTYSTDRLRK; (SEQ ID NO: 28)
    GSGTYSTDRLRKWMF; (SEQ ID NO: 29)
    VLTYSTDRLRKWMFI; (SEQ ID NO: 30)
    HTAIVLTYSTDRLR; (SEQ ID NO: 31)
    LTYSTDRLRKWMFI; (SEQ ID NO: 32)
    TYSTDRLRKWMFI; (SEQ ID NO: 33)
    HTAIVLTYSTDR; (SEQ ID NO: 34)
    GSGTYDRLRKWM; (SEQ ID NO: 35)
    GSGTYSTDRLRK; (SEQ ID NO: 36)
    GSTDRLRKWMFI; (SEQ ID NO: 37)
    GSTDRLRKWMFA; (SEQ ID NO: 38)
    GSTDRLRKWMAI; (SEQ ID NO: 39)
    GSTDRLRKWAFI; (SEQ ID NO: 40)
    GSTDRLRKAMFI; (SEQ ID NO: 41)
    GSTDRLRKAXFI; (SEQ ID NO: 42)
    YSTDRLRKWMFI; (SEQ ID NO: 43)
    GSTDRLRKMFI; (SEQ ID NO: 44)
    GSGRLRKWMFI; (SEQ ID NO: 45)
    STDRLRKWMFI; (SEQ ID NO: 46)
    YSTDRLRKWMF; (SEQ ID NO: 47)
    STDRLRKWMF; (SEQ ID NO: 48)
    ADRLRKWMFI; (SEQ ID NO: 49)
    GSRLRKWMFI; (SEQ ID NO: 50)
    TDRLRKWMFI; (SEQ ID NO: 51)
    TDRLRKWXFI; (SEQ ID NO: 52)
    TARLRKWMFI; (SEQ ID NO: 53)
    TDALRKWMFI; (SEQ ID NO: 54)
    TDRARKWMFI; (SEQ ID NO: 55)
    TDRLAKWMFI; (SEQ ID NO: 56)
    TDRLRAWMFI; (SEQ ID NO: 57)
    TDRLRKAMFI; (SEQ ID NO: 58)
    TDRLRKWAFI; (SEQ ID NO: 59)
    TDRLRKWMAI; (SEQ ID NO: 60)
    TDRLRKWMFA; (SEQ ID NO: 61)
    TYSTDRLRK; (SEQ ID NO: 62)
    STDRLRKMF; (SEQ ID NO: 63)
    GSGRLRKWM; (SEQ ID NO: 64)
    GRLRKWMFI; (SEQ ID NO: 65)
    DRLRKWMFI; (SEQ ID NO: 66)
    DRLRKWMAI; (SEQ ID NO: 67)
    GSTDRLRK; (SEQ ID NO: 68)
    RLRKWMFI; (SEQ ID NO: 69)
    and
    RLRKWMAI. (SEQ ID NO: 70)
  • In a fifth aspect, P comprises at least 3 contiguous amino acids of the i3 loop.
  • In a specific embodiment of the fifth aspect, P is derived from the i3 loop and is represented by the following structural formula or a pharmaceutically acceptable salt thereof:
  • Z1-Z2-Z3-Z4-Z5-Z6-Z7-Z8-Z9-Z10-Z11-Z12-Z13-Z14-Z15-Z16-
    Z17-Z18-Z19-Z20-Z21-Z22-Z23-Z24-Z25-Z26-Z27-Z28-Z29-Z30-R1,
  • wherein:
  • Z1 is absent or a phenylalanine residue;
  • Z2 is absent or an asparagine residue;
  • Z3 is absent or an isoleucine residue;
  • Z4 is absent or a valine residue;
  • Z5 is absent or an arginine residue;
  • Z6 is absent or an isoleucine residue;
  • Z7 is absent or a leucine residue;
  • Z8 is absent or a methionine residue;
  • Z9 is absent or a threonine residue;
  • Z10 is absent or a lysine residue;
  • Z11 is absent or a leucine residue;
  • Z12 is absent or an arginine residue;
  • Z13 is absent or an alanine residue;
  • Z14 is absent or a serine residue;
  • Z15 is absent or a threonine residue;
  • Z16 is absent or a threonine residue;
  • Z17 is absent or a serine residue;
  • Z18 is absent or a glutamic acid residue;
  • Z19 is absent or a threonine residue;
  • Z20 is absent or an isoleucine residue;
  • Z21 is absent or a glutamine residue;
  • Z22 is absent or a tyrosine residue;
  • Z23 is absent or an arginine residue;
  • Z24 is absent or a lysine residue;
  • Z25 is absent or an alanine residue;
  • Z26 is absent or a valine residue;
  • Z27 is absent or a lysine residue;
  • Z28 is absent or an alanine or serine residue;
  • Z29 is absent or a threonine residue; and
  • Z30 is absent or a leucine residue;
  • provided that at least five of Z1-Z30 are present;
  • R1 is OR2 or N(R2)2;
  • each R2 is independently hydrogen or (C1-C10)alkyl; and
  • from 0 to 5 amino acid residues are present in the D configuration.
  • In a specific embodiment, at least three of Z5, Z6, Z7, Z8, and Z9 are present.
  • In a further specific embodiment,
  • Z5 is an arginine residue;
  • Z6 is an isoleucine residue; and
  • Z7 is a leucine residue.
  • In another further specific embodiment,
  • Z7 is a leucine residue;
  • Z8 is a methionine residue; and
  • Z9 is a threonine residue.
  • In a another specific embodiment,
  • Z5 is an arginine residue;
  • Z6 is an isoleucine residue;
  • Z7 is a leucine residue; and
  • Z8 is a methionine residue.
  • In a another specific embodiment,
  • Z6 is an isoleucine residue;
  • Z7 is a leucine residue;
  • Z8 is absent or a methionine residue; and
  • Z9 is a threonine residue.
  • In a another specific embodiment,
  • Z5 is an arginine residue;
  • Z6 is an isoleucine residue;
  • Z7 is a leucine residue;
  • Z8 is a methionine residue; and
  • Z9 is a threonine residue.
  • In yet another specific embodiment, at least three of Z10, Z11, Z12, and Z13 are present.
  • In a another specific embodiment,
  • Z10 is a lysine residue;
  • Z11 is a leucine residue; and
  • Z12 is an arginine residue.
  • In a another specific embodiment,
  • Z11 is a leucine residue;
  • Z12 is an arginine residue; and
  • Z13 is an alanine residue.
  • In yet another specific embodiment,
  • Z10 is a lysine residue;
  • Z11 is a leucine residue;
  • Z12 is an arginine residue; and
  • Z13 is an alanine residue.
  • In another specific embodiment, at least three of Z14, Z15, Z16, and Z17 are present.
  • In another specific embodiment,
  • Z14 is a serine residue;
  • Z15 is a threonine residue; and
  • Z16 is a threonine residue.
  • In another specific embodiment,
  • Z15 is a threonine residue;
  • Z16 is a threonine residue; and
  • Z17 is a serine residue;
  • In another specific embodiment,
  • Z14 is a serine residue;
  • Z15 is a threonine residue;
  • Z16 is a threonine residue; and
  • Z17 is a serine residue;
  • In another specific embodiment, at least three of Z19, Z20, Z21, Z22, and Z23 are present.
  • In another specific embodiment,
  • Z19 is a threonine residue;
  • Z20 is an isoleucine residue; and
  • Z21 is a glutamine residue.
  • In another specific embodiment,
  • Z21 is a glutamine residue;
  • Z22 is a tyrosine residue; and
  • Z23 is an arginine residue.
  • In another specific embodiment,
  • Z19 is a threonine residue;
  • Z20 is an isoleucine residue;
  • Z21 is a glutamine residue; and
  • Z22 is a tyrosine residue.
  • In another specific embodiment,
  • Z20 is an isoleucine residue;
  • Z21 is a glutamine residue;
  • Z22 is a tyrosine residue; and
  • Z23 is an arginine residue.
  • In another specific embodiment,
  • Z19 is a threonine residue;
  • Z20 is an isoleucine residue;
  • Z21 is a glutamine residue;
  • Z22 is a tyrosine residue; and
  • Z23 is an arginine residue.
  • In another specific embodiment, at least three of Z24, Z25, Z26, Z27, and Z28 are present.
  • In another specific embodiment,
  • Z24 is a lysine residue;
  • Z25 is an alanine residue; and
  • Z26 is a valine residue.
  • In another specific embodiment,
  • Z26 is a valine residue;
  • Z27 is a lysine residue; and
  • Z28 is an alanine or serine residue.
  • In another specific embodiment,
  • Z24 is a lysine residue;
  • Z25 is an alanine residue;
  • Z26 is a valine residue; and
  • Z27 is a lysine residue.
  • In another specific embodiment,
  • Z25 is an alanine residue;
  • Z26 is a valine residue;
  • Z27 is a lysine residue; and
  • Z28 is an alanine or serine residue.
  • In another specific embodiment,
  • Z24 is a lysine residue;
  • Z25 is an alanine residue;
  • Z26 is a valine residue;
  • Z27 is a lysine residue; and
  • Z28 is an alanine or serine residue. Specifically, Z28 is an alanine residue.
  • In a specific embodiment of the fifth aspect, the i3 loop of the CRF1 receptor from which P is derived has the following sequence: FNIVRILMTKLRASTTSETIQYRKAVKA (SEQ ID NO: 71).
  • In another embodiment of the fifth aspect, P is a sequence selected from:
  • FNIVRILMTKLRASTTSETIQYRKAVKA; (SEQ ID NO: 71)
    IVRILMTKLRASTTSETIQYRKAVKA; (SEQ ID NO: 72)
    FNIVRILMTKLRASTTSETIQYRKAV; (SEQ ID NO: 73)
    RILMTKLRASTTSETIQYRKAVKA; (SEQ ID NO: 74)
    FNIVRILMTKLRASTTSETIQYRK; (SEQ ID NO: 75)
    FNIVRILMTKLRASTTSETIQYR; (SEQ ID NO: 76)
    LMTKLRASTTSETIQYRKAVKA; (SEQ ID NO: 77)
    FNIVRILMTKLRASTTSETIQY; (SEQ ID NO: 78)
    TKLRASTTSETIQYRKAVKA; (SEQ ID NO: 79)
    LMTKLRASTTSETIQYRKAV; (SEQ ID NO: 80)
    LRASTTSETIQYRKAVKA; (SEQ ID NO: 81)
    LMTKLRASTTSETIQYRK; (SEQ ID NO: 82)
    LMTKLRASTTSETIQYR; (SEQ ID NO: 83)
    ASTTSETIQYRKAVKA; (SEQ ID NO: 84)
    ASTTSETIQYRKAVKS; (SEQ ID NO: 85)
    LMTKLRASTTSETIQ; (SEQ ID NO: 86)
    SETIQYRKAVKATL; (SEQ ID NO: 87)
    LMTKLRASTTSETI; (SEQ ID NO: 88)
    RILMTKLRASTTS; (SEQ ID NO: 89)
    SETIQYRKAVKA; (SEQ ID NO: 90)
    TKLRASTTSETI; (SEQ ID NO: 91)
    ASTTSETIQYR; (SEQ ID NO: 92)
    TKLRASTTSET; (SEQ ID NO: 93)
    KLRASTTSETI; (SEQ ID NO: 94)
    TIQYRKAVKA; (SEQ ID NO: 95)
    KLRASTTSET; (SEQ ID NO: 96)
    RILMTKLRA; (SEQ ID NO: 97)
    and
    KLRASTTSE. (SEQ ID NO: 98)
  • In a sixth aspect, P comprises at least 3 contiguous amino acids of the i4 domain.
  • In a specific embodiment of the sixth aspect, P derived from the i4 domain and is represented by the following structural formula or a pharmaceutically acceptable salt thereof,
  • M1-M2-M3-M4-M5-M6-M7-M8-M9-M10-M11-M12-M13-M14-M15-M16-
    M17-M18-M19-M20-M21-M22-M23-M24-M25-M26-M27-M28-M29-M30-
    M31-M32-M33-M34-M35-M36-M37-M38-M39-M40-M41-M42-M43-M44-
    M45-M46-M47-R1,
  • wherein:
  • M1 is absent or a glutamic acid residue;
  • M2 is absent or a valine residue;
  • M3 is absent or an arginine residue;
  • M4 is absent or a serine residue;
  • M5 is absent or an alanine residue;
  • M6 is absent or an isoleucine residue;
  • M7 is absent or an arginine residue;
  • M8 is absent or a lysine residue;
  • M9 is absent or an arginine residue;
  • M10 is absent or a tryptophan residue;
  • M11 is absent or a histidine residue;
  • M12 is absent or an arginine residue;
  • M13 is absent or a tryptophan residue;
  • M14 is absent or a glutamine residue;
  • M15 is absent or an aspartic acid residue;
  • M16 is absent or a lysine residue;
  • M17 is absent or a histidine or glycine residue;
  • M18 is absent or a serine residue;
  • M19 is absent or an isoleucine residue;
  • M20 is absent or an arginine residue;
  • M21 is absent or an alanine residue;
  • M22 is absent or an arginine residue;
  • M23 is absent or a valine residue;
  • M24 is absent or an alanine residue;
  • M25 is absent or an arginine residue;
  • M26 is absent or an alanine residue;
  • M27 is absent or a methionine residue;
  • M28 is absent or a serine residue;
  • M29 is absent or an isoleucine residue;
  • M30 is absent or a proline residue;
  • M31 is absent or a threonine residue;
  • M32 is absent or a serine residue;
  • M33 is absent or a proline residue;
  • M34 is absent or a threonine residue;
  • M35 is absent or an arginine residue;
  • M36 is absent or a valine residue;
  • M37 is absent or a serine residue;
  • M38 is absent or a phenylalanine residue;
  • M39 is absent or a histidine residue;
  • M40 is absent or a serine residue;
  • M41 is absent or an isoleucine residue;
  • M42 is absent or a lysine residue;
  • M43 is absent or a glutamine residue;
  • M44 is absent or a serine residue;
  • M45 is absent or a threonine residue;
  • M46 is absent or an alanine residue; and
  • M47 is absent or a valine residue; provided that at least five of M1-M47 are present;
  • R1 is OR2 or N(R2)2;
  • each R2 is independently hydrogen or (C1-C10)alkyl; and
  • from 0 to 5 amino acid residues are present in the D configuration.
  • It is understood that when P is described as M1-M47 it is bonded to L as written. For example, M1 is bonded to L. If M1 is absent, then M2 is bonded to L.
  • In a specific embodiment, at least three of M2-M7 are present.
  • In another specific embodiment,
  • M2 is a valine residue;
  • M3 is an arginine residue; and
  • M4 is a serine residue.
  • In another specific embodiment,
  • M5 is an alanine residue;
  • M6 is an isoleucine residue; and
  • M7 is an arginine residue.
  • In another specific embodiment,
  • M2 is a valine residue;
  • M3 is an arginine residue;
  • M4 is a serine residue; and
  • M5 is an alanine residue.
  • In another specific embodiment,
  • M4 is a serine residue;
  • M5 is an alanine residue;
  • M6 is an isoleucine residue; and
  • M7 is an arginine residue.
  • In another specific embodiment,
  • M2 is a valine residue;
  • M3 is an arginine residue;
  • M4 is a serine residue;
  • M5 is an alanine residue;
  • M6 is an isoleucine residue.
  • In another specific embodiment,
  • M3 is an arginine residue;
  • M4 is a serine residue;
  • M5 is an alanine residue;
  • M6 is an isoleucine residue; and
  • M7 is an arginine residue.
  • In another specific embodiment,
  • M2 is a valine residue;
  • M3 is an arginine residue;
  • M4 is a serine residue;
  • M5 is an alanine residue;
  • M6 is an isoleucine residue; and
  • M7 is an arginine residue.
  • In another specific embodiment, at least three of M9-M13 are present.
  • In another specific embodiment,
  • M9 is an arginine residue;
  • M10 is a tryptophan residue;
  • M11 is a histidine residue;
  • M12 is an arginine residue.
  • In another specific embodiment,
  • M10 is a tryptophan residue;
  • M11 is a histidine residue;
  • M12 is an arginine residue; and
  • M13 is a tryptophan residue.
  • In another specific embodiment,
  • M9 is an arginine residue;
  • M10 is a tryptophan residue;
  • M11 is a histidine residue;
  • M12 is an arginine residue; and
  • M13 is a tryptophan residue.
  • In another specific embodiment, at least three of M18-M22 are present.
  • In another specific embodiment,
  • M18 is a serine residue;
  • M19 is an isoleucine residue;
  • M20 is an arginine residue; and
  • M21 is an alanine residue.
  • In another specific embodiment,
  • M19 is an isoleucine residue;
  • M20 is an arginine residue;
  • M21 is an alanine residue; and
  • M22 is an arginine residue.
  • In another specific embodiment,
  • M18 is a serine residue;
  • M19 is an isoleucine residue;
  • M20 is an arginine residue;
  • M21 is an alanine residue; and
  • M22 is an arginine residue.
  • In another specific embodiment, at least three of M23-M27 are present.
  • In another specific embodiment,
  • M23 is a valine residue;
  • M24 is an alanine residue;
  • M25 is an arginine residue; and
  • M26 is an alanine residue.
  • In another specific embodiment,
  • M24 is an alanine residue;
  • M25 is an arginine residue;
  • M26 is an alanine residue;
  • M27 is a methionine residue.
  • In another specific embodiment,
  • M23 is a valine residue;
  • M24 is an alanine residue;
  • M25 is an arginine residue;
  • M26 is an alanine residue;
  • M27 is a methionine residue.
  • In another specific embodiment, at least three of M25-M29 are present.
  • In another specific embodiment,
  • M25 is an arginine residue;
  • M26 is an alanine residue;
  • M27 is a methionine residue; and
  • M28 is a serine residue.
  • In another specific embodiment,
  • M26 is an alanine residue;
  • M27 is a methionine residue;
  • M28 is a serine residue; and
  • M29 is an isoleucine residue.
  • In another specific embodiment,
  • M25 is an arginine residue;
  • M26 is an alanine residue;
  • M27 is a methionine residue;
  • M28 is a serine residue; and
  • M29 is an isoleucine residue.
  • In another specific embodiment, at least three of M32-M36 are present.
  • In another specific embodiment,
  • M32 is a serine residue;
  • M33 is a proline residue;
  • M34 is a threonine residue; and
  • M35 is an arginine residue.
  • In another specific embodiment,
  • M33 is a proline residue;
  • M34 is a threonine residue;
  • M35 is an arginine residue; and
  • M36 is a valine residue.
  • In another specific embodiment,
  • M32 is a serine residue;
  • M33 is a proline residue;
  • M34 is a threonine residue;
  • M35 is an arginine residue; and
  • M36 is a valine residue.
  • In a specific embodiment of the sixth aspect, the i4 domain of the CRF1 receptor from which P is derived has the following sequence:
  • (SEQ ID NO: 99)
    EVRSAIRKRWHRWQDKHSIRARVARAMSIPTSPTRVSFHSIKQSTAV.
  • In another embodiment of the sixth aspect, P is a sequence selected from:
  • RAMSIPTSPTRVSFHSIKQSTAV; (SEQ ID NO: 100)
    HSIRARVARAMSIPTSPTRV; (SEQ ID NO: 101)
    RWQDKHSIRARVARAMSI; (SEQ ID NO: 102)
    EVRSAIRKRWHRWQD; (SEQ ID NO: 103)
    RWHRWQDKHSIRAR; (SEQ ID NO: 104)
    VRSAIRKRWHRW; (SEQ ID NO: 105)
    and
    GSIRARVARAM. (SEQ ID NO: 106)
  • In a seventh aspect, T is an optionally substituted (C6-C30)alkyl, (C6-C30)alkenyl, (C6-C30)alkynyl, wherein 0-3 carbon atoms are replaced with oxygen, sulfur, nitrogen or a combination thereof. This value of T is applicable to the first, second, third, fourth, fifth and sixth aspects and the specific (i.e., specific, more specific and most specific) embodiments of same.
  • In a specific embodiment of the seventh aspect, T is selected from: CH3(CH2)16, CH3(CH2)15, CH3(CH2)14, CH3(CH2)13, CH3(CH2)12, CH3(CH2)11, CH3(CH2)10, CH3(CH2)9, CH3(CH2)8, CH3(CH2)9OPh-, CH3(CH2)6C═C(CH2)6, CH3(CH2)11O(CH2)3, and CH3(CH2)9O(CH2)2.
  • In another specific embodiment of the seventh aspect, T is a fatty acid derivative.
  • In a more specific embodiment of the seventh aspect, the fatty acid is selected from the group consisting of: butyric acid, caproic acid, caprylic acid, capric acid, lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, lignoceric acid, myristoleic acid, palmitoleic acid, oleic acid, linoleic acid, α-linolenic acid, arachidonic acid, eicosapentaenoic acid, erucic acid, docosahexaenoic acid.
  • In an eighth aspect, T is a bile acid derivative. This value of T is applicable to the first, second, third, fourth, fifth and sixth aspects and the specific (i.e., specific, more specific and most specific) embodiments of same.
  • In a specific embodiment of the eighth aspect, the bile acid is selected from the group consisting of: lithocholic acid, chenodeoxycholic acid, deoxycholic acid, cholanic acid, cholic acid, ursocholic acid, ursodeoxycholic acid, isoursodeoxycholic acid, lagodeoxycholic acid, dehydrocholic acid, hyocholic acid, and hyodeoxycholic acid.
  • In a ninth aspect, T is selected from sterols; progestagens; glucocorticoids; mineralcorticoids; androgens; and estrogens. This value of T is applicable to the first, second, third, fourth, fifth and sixth aspects and the specific (i.e., specific, more specific and most specific) embodiments of same.
  • In a tenth aspect, T-L of Formula I is represented by a moiety selected from the group consisting of:
      • CH3(CH2)15—C(O);
      • CH3(CH2)13—C(O);
      • CH3(CH2)9O(CH2)2C(O);
      • CH3(CH2)10O(CH2)2C(O);
      • CH3(CH2)6C═C(CH2)6—C(O);
      • LCA-C(O); and
      • CH3(CH2)9OPh-C(O) wherein
  • Figure US20110301087A1-20111208-C00028
  • In an eleventh aspect, T of Formula I is represented by a moiety selected from the group consisting of:
  • Figure US20110301087A1-20111208-C00029
  • and
  • Figure US20110301087A1-20111208-C00030
  • In yet another embodiment, a GPCR compound of the invention is selected from one of the following compounds or a pharmaceutically acceptable salt thereof:
  • Figure US20110301087A1-20111208-C00031
    Figure US20110301087A1-20111208-C00032
    Figure US20110301087A1-20111208-C00033
    Figure US20110301087A1-20111208-C00034
    Figure US20110301087A1-20111208-C00035
    Figure US20110301087A1-20111208-C00036
  • In the above listing of compounds, the structure follows the number identifier.
  • In yet another embodiment, a GPCR compound of the invention is selected from one of the following compounds or a pharmaceutically acceptable salt thereof:
  • Figure US20110301087A1-20111208-C00037
    Figure US20110301087A1-20111208-C00038
    Figure US20110301087A1-20111208-C00039
    Figure US20110301087A1-20111208-C00040
    Figure US20110301087A1-20111208-C00041
    Figure US20110301087A1-20111208-C00042
    Figure US20110301087A1-20111208-C00043
    Figure US20110301087A1-20111208-C00044
    Figure US20110301087A1-20111208-C00045
    Figure US20110301087A1-20111208-C00046
    Figure US20110301087A1-20111208-C00047
    Figure US20110301087A1-20111208-C00048
    Figure US20110301087A1-20111208-C00049
    Figure US20110301087A1-20111208-C00050
    Figure US20110301087A1-20111208-C00051
    Figure US20110301087A1-20111208-C00052
    Figure US20110301087A1-20111208-C00053
    Figure US20110301087A1-20111208-C00054
    Figure US20110301087A1-20111208-C00055
    Figure US20110301087A1-20111208-C00056
  • In the above listing of compounds, structures follow the compound number identifier.
  • In yet another embodiment, a GPCR compound of the invention is selected from one of the following compounds or a pharmaceutically acceptable salt thereof:
  • Figure US20110301087A1-20111208-C00057
    Figure US20110301087A1-20111208-C00058
    Figure US20110301087A1-20111208-C00059
    Figure US20110301087A1-20111208-C00060
  • In the above listing of compounds the structure follows the compound number identifier.
  • In yet another embodiment, a GPCR compound of the invention is selected from one of the following compounds or a pharmaceutically acceptable salt thereof:
  • Figure US20110301087A1-20111208-C00061
  • In the above listing of compounds, the structure follows the number identifier.
  • “Cycloalkyl” used alone or as part of a larger moiety such as “cycloalkylalkyl” refers to a monocyclic or polycyclic, non-aromatic ring system of 3 to 20 carbon atoms, 3 to 12 carbon atoms, or 3 to 9 carbon atoms, which may be saturated or unsaturated. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cyclohexa-1,3-dienyl, cyclooctyl, cycloheptanyl, norbornyl, adamantyl, and the like.
  • “Heterocycloalkyl” refers to a saturated or unsaturated, non-aromatic, monocyclic or polycyclic ring system of 3 to 20 atoms, 3 to 12 atoms, or 3 to 8 atoms, containing one to four ring heteroatoms chosen from O, N and S. Examples of heterocyclyl groups include pyrrolidine, piperidine, tetrahydrofuran, tetrahydropyran, tetrahydrothiophene, tetrahydrothiopyran, isoxazolidine, 1,3-dioxolane, 1,3-dithiolane, 1,3-dioxane, 1,4-dioxane, 1,3-dithiane, 1,4-dithiane, morpholine, thiomorpholine, thiomorpholine-1,1-dioxide, tetrahydro-2H-1,2-thiazine-1,1-dioxide, isothiazolidine-1,1-dioxide, pyrrolidin-2-one, piperidin-2-one, piperazin-2-one, and morpholin-2-one, and the like.
  • “Halogen” and “halo” refer to fluoro, chloro, bromo or iodo.
  • “Haloalkyl” refers to an alkyl group substituted with one or more halogen atoms. By analogy, “haloalkenyl”, “haloalkynyl”, etc., refers to the group (for example alkenyl or alkynyl) substituted by one or more halogen atoms.
  • “Cyano” refers to the group —CN.
  • “Oxo” refers to a divalent ═O group.
  • “Thioxo” refers to a divalent ═S group.
  • “Ph” refers to a phenyl group.
  • “Carbonyl” refers to a divalent —C(O)— group.
  • “Alkyl” used alone or as part of a larger moiety such as “hydroxyalkyl”, “alkoxyalkyl”, “alkylamine” refers to a straight or branched, saturated aliphatic group having the specified number of carbons, typically having 1 to 12 carbon atoms. More particularly, the aliphatic group may have 1 to 10, 1 to 8, 1 to 6, or 1 to 4 carbon atoms. This term is exemplified by groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-hexyl, and the like.
  • “Alkenyl” refers to a straight or branched aliphatic group with at least one double bond. Typically, alkenyl groups have from 2 to 12 carbon atoms, from 2 to 8, from 2 to 6, or from 2 to 4 carbon atoms. Examples of alkenyl groups include ethenyl (—CH═CH2), n-2-propenyl (allyl, —CH2CH═CH2), pentenyl, hexenyl, and the like.
  • “Alkynyl” refers to a straight or branched aliphatic group having at least 1 site of alkynyl unsaturation. Typically, alkynyl groups contain 2 to 12, 2 to 8, 2 to 6 or 2 to 4 carbon atoms. Examples of alkynyl groups include ethynyl (—C≡CH), propargyl (—CH2C≡CH), pentynyl, hexynyl, and the like.
  • “Alkylene” refers to a bivalent saturated straight-chained hydrocarbon, e.g., C1-C6 alkylene includes —(CH2)6—, —CH2—CH—(CH2)3CH3, and the like. “Bivalent means that the alkylene group is attached to the remainder of the molecule through two different carbon atoms.
  • “Alkenylene” refers to an alkylene group with in which one carbon-carbon single bond is replaced with a double bond.
  • “Alkynylene” refers to an alkylene group with in which one carbon-carbon single bond is replaced with a triple bond.
  • “Aryl” used alone or as part of a larger moiety as in “aralkyl” refers to an aromatic carbocyclic group of from 6 to 14 carbon atoms having a single ring or multiple condensed rings. The term “aryl” also includes aromatic carbocycle(s) fused to cycloalkyl or heterocycloalkyl groups. Examples of aryl groups include phenyl, benzo[d][1,3]dioxole, naphthyl, phenantrenyl, and the like.
  • “Aryloxy” refers to an —OAr group, wherein 0 is an oxygen atom and Ar is an aryl group as defined above.
  • “Aralkyl” refers to an alkyl having at least one alkyl hydrogen atom replaced with an aryl moiety, such as benzyl, —(CH2)2phenyl, —(CH2)3phenyl, —CH(phenyl)2, and the like.
  • “Alkyl cycloalkyl” refers to an alkyl having at least one alkyl hydrogen atom replaced with a cycloalkyl moiety, such as —CH2-cyclohexyl, —CH2-cyclohexenyl, and the like.
  • “Heteroaryl” used alone or a part of a larger moiety as in “heteroaralkyl” refers to a 5 to 14 membered monocyclic, bicyclic or tricyclic heteroaromatic ring system, containing one to four ring heteroatoms independently selected from nitrogen, oxygen and sulfur. The term “heteroaryl” also includes heteroaromatic ring(s) fused to cycloalkyl or heterocycloalkyl groups. Particular examples of heteroaryl groups include optionally substituted pyridyl, pyrrolyl, pyrimidinyl, furyl, thienyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyrazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl,1,3,4-triazinyl, 1,2,3-triazinyl, benzofuryl, [2,3-dihydro]benzofuryl, isobenzofuryl, benzothienyl, benzotriazolyl, isobenzothienyl, indolyl, isoindolyl, 3H-indolyl, benzimidazolyl, imidazo[1,2-a]pyridyl, benzothiazolyl, benzoxazolyl, quinolizinyl, quinazolinyl, pthalazinyl, quinoxalinyl, cinnolinyl, napthyridinyl, pyrido[3,4-b]pyridyl, pyrido[3,2-b]pyridyl, pyrido[4,3-b]pyridyl, quinolyl, isoquinolyl, tetrazolyl, 1,2,3,4-tetrahydroquinolyl, 1,2,3,4-tetrahydroisoquinolyl, purinyl, pteridinyl, carbazolyl, xanthenyl, benzoquinolyl, and the like.
  • “Heteroaryloxy” refers to an —OHet group, wherein 0 is an oxygen atom and Het is a heteroaryl group as defined above.
  • “Heteroaralkyl” refers to an alkyl having at least one alkyl hydrogen atom replaced with a heteroaryl moiety, such as —CH2-pyridinyl, —CH2-pyrimidinyl, and the like.
  • “Alkoxy” refers to the group —O—R where R is “alkyl”, “cycloalkyl”, “alkenyl”, or “alkynyl”. Examples of alkoxy groups include for example, methoxy, ethoxy, ethenoxy, and the like.
  • “Alkyl heterocycloalkyl” refers to an alkyl having at least one alkyl hydrogen atom replaced with a heterocycloalkyl moiety, such as —CH2-morpholino, —CH2-piperidyl and the like.
  • “Alkoxycarbonyl” refers to the group —C(O)OR where R is “alkyl”, “alkenyl”, “alkynyl”, “cycloalkyl”, “heterocycloalkyl”, “aryl”, or “heteroaryl”.
  • “Hydroxyalkyl” and “alkoxyalkyl” are alky groups substituted with hydroxyl and alkoxy, respectively.
  • “Amino” means —NH2; “alkylamine” and “dialkylamine” mean —NHR and —NR2, respectively, wherein R is an alkyl group. “Cycloalkylamine” and “dicycloalkylamine” mean —NHR and —NR2, respectively, wherein R is a cycloalkyl group. “Cycloalkylalkylamine” means —NHR wherein R is a cycloalkylalkyl group. “[Cycloalkylalkyl][alkyl]amine” means —N(R)2 wherein one R is cycloalkylalkyl and the other R is alkyl.
  • Haloalkyl and halocycloalkyl include mono, poly, and perhaloalkyl groups where the halogens are independently selected from fluorine, chlorine, bromine and iodine.
  • Suitable substituents for “alkyl”, “alkenyl”, “alkynyl”, “cycloalkyl”, “heterocycloalkyl”, “aryl”, or “heteroaryl”, etc., are those which will form a stable compound of the invention. Examples of suitable substituents are those selected from the group consisting of halogen, —CN, —OH, —NH2, (C1-C4)alkyl, (C1-C4)haloalkyl, aryl, heteroaryl, (C3-C7)cycloalkyl, (5-7 membered) heterocycloalkyl, —NH(C1-C6)alkyl, —N((C1-C6)alkyl)2, (C1-C6)alkoxy, (C1-C6)alkoxycarbonyl, —CONH2, —OCONH2, —NHCONH2, —N(C1-C6)alkylCONH2, —N(C1-C6)alkylCONH(C1-C6)alkyl, —NHCONH(C1-C6)alkyl, —NHCON((C1-C6)alkyl)2, —N(C1-C6)alkylCON((C1-C6)alkyl)2, —NHC(S)NH2, —N(C1-C6)alkylC(S)NH2, —N(C1-C6)alkylC(S)NH(C1-C6)alkyl, —NHC(S)NH(C1-C6)alkyl, —NHC(S)N((C1-C6)alkyl)2, —N(C1-C6)alkylC(S)N((C1-C6)alkyl)2, —CONH(C1-C6)alkyl, —OCONH(C1-C6)alkyl —CON((C1-C6)alkyl)2, —C(S)(C1-C6)alkyl, —S(O)p(C1-C6)alkyl, —S(O)pNH2, —S(O)pNH(C1-C6)alkyl, —S(O)pN((C1-C6)alkyl)2, —CO(C1-C6)alkyl, —OCO(C1-C6)alkyl, —C(O)O(C1-C6)alkyl, —OC(O)O(C1-C6)alkyl, —C(O)H or —CO2H. More particularly, the substituents are selected from halogen, —CN, —OH, —NH2, (C1-C4)alkyl, (C1-C4)haloalkyl, (C1-C4)alkoxy, phenyl, and (C3-C7)cycloalkyl. Within the framework of this invention, said “substitution” is also meant to encompass situations where a hydrogen atom is replaced with a deuterium atom. p is an integer with a value of 1 or 2.
  • Pharmaceutically acceptable salts of the compounds disclosed herein are included in the present invention. For example, an acid salt of a compound containing an amine or other basic group can be obtained by reacting the compound with a suitable organic or inorganic acid, resulting in pharmaceutically acceptable anionic salt forms. Examples of anionic salts include the acetate, benzenesulfonate, benzoate, bicarbonate, bitartrate, bromide, calcium edetate, camsylate, carbonate, chloride, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, glyceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, malate, maleate, mandelate, mesylate, methylsulfate, mucate, napsylate, nitrate, pamoate, pantothenate, phosphate/diphospate, polygalacturonate, salicylate, stearate, subacetate, succinate, sulfate, tannate, tartrate, teoclate, tosylate, and triethiodide salts.
  • Salts of the compounds containing an acidic functional group can be prepared by reacting with a suitable base. Such a pharmaceutically acceptable salt can be made with a base which affords a pharmaceutically acceptable cation, which includes alkali metal salts (especially sodium and potassium), alkaline earth metal salts (especially calcium and magnesium), aluminum salts and ammonium salts, as well as salts made from physiologically acceptable organic bases such as trimethylamine, triethylamine, morpholine, pyridine, piperidine, picoline, dicyclohexylamine, N,N′-dibenzylethylenediamine, 2-hydroxyethylamine, bis-(2-hydroxyethyl)amine, tri-(2-hydroxyethyl)amine, procaine, dibenzylpiperidine, dehydroabietylamine, N,N′-bisdehydroabietylamine, glucamine, N-methylglucamine, collidine, quinine, quinoline, and basic amino acids such as lysine and arginine.
  • Pharmaceutical Compositions
  • The invention also provides pharmaceutical compositions comprising an effective amount of a compound Formula I (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt of said compound; and a pharmaceutically acceptable carrier. The carrier(s) are “pharmaceuticallyacceptable” in that they are not deleterious to the recipient thereof in an amount used in the medicament.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • If required, the solubility and bioavailability of the compounds of the present invention in pharmaceutical compositions may be enhanced by methods well-known in the art. One method includes the use of lipid excipients in the formulation. See “Oral Lipid-Based Formulations: Enhancing the Bioavailability of Poorly Water-Soluble Drugs (Drugs and the Pharmaceutical Sciences),” David J. Hauss, ed. Informa Healthcare, 2007; and “Role of Lipid Excipients in Modifying Oral and Parenteral Drug Delivery: Basic Principles and Biological Examples,” Kishor M. Wasan, ed. Wiley-Interscience, 2006.
  • Another known method of enhancing bioavailability is the use of an amorphous form of a compound of this invention optionally formulated with a poloxamer, such as LUTROL™ and PLURONIC™ (BASF Corporation), or block copolymers of ethylene oxide and propylene oxide. See U.S. Pat. No. 7,014,866; and United States patent publications 20060094744 and 20060079502.
  • The pharmaceutical compositions of the invention include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), pulmonary, vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration. In certain embodiments, the compound of the formulae herein is administered transdermally (e.g., using a transdermal patch or iontophoretic techniques). Other formulations may conveniently be presented in unit dosage form, e.g., tablets, sustained release capsules, and in liposomes, and may be prepared by any methods well known in the art of pharmacy. See, for example, Remington's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, Pa. (17th ed. 1985).
  • Such preparative methods include the step of bringing into association with the molecule to be administered ingredients such as the carrier that constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • In certain embodiments, the compound is administered orally. Compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets, or tablets each containing a predetermined amount of the active ingredient; a powder or granules; a solution or a suspension in an aqueous liquid or a non-aqueous liquid; an oil-in-water liquid emulsion; a water-in-oil liquid emulsion; packed in liposomes; or as a bolus, etc. Soft gelatin capsules can be useful for containing such suspensions, which may beneficially increase the rate of compound absorption.
  • In the case of tablets for oral use, carriers that are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • Compositions suitable for oral administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; and pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia.
  • Compositions suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • Such injection solutions may be in the form, for example, of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant.
  • The pharmaceutical compositions of this invention may be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • The pharmaceutical compositions of this invention may be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art. See, e.g.: Rabinowitz J D and Zaffaroni A C, U.S. Pat. No. 6,803,031, assigned to Alexza Molecular Delivery Corporation.
  • Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application. For topical application topically to the skin, the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax, and water. Alternatively, the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol, and water. The pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches and iontophoretic administration are also included in this invention.
  • Application of the patient therapeutics may be local, so as to be administered at the site of interest. Various techniques can be used for providing the patient compositions at the site of interest, such as injection, use of catheters, trocars, projectiles, pluronic gel, stents, sustained drug release polymers or other device which provides for internal access.
  • Thus, according to yet another embodiment, the compounds of this invention may be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents, or catheters. Suitable coatings and the general preparation of coated implantable devices are known in the art and are exemplified in U.S. Pat. Nos. 6,099,562; 5,886,026; and 5,304,121. The coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof. The coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysaccharides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics in the composition. Coatings for invasive devices are to be included within the definition of pharmaceutically acceptable carrier, adjuvant or vehicle, as those terms are used herein.
  • According to another embodiment, the invention provides a method of coating an implantable medical device comprising the step of contacting said device with the coating composition described above. It will be obvious to those skilled in the art that the coating of the device will occur prior to implantation into a mammal.
  • According to another embodiment, the invention provides a method of impregnating an implantable drug release device comprising the step of contacting said drug release device with a compound or composition of this invention. Implantable drug release devices include, but are not limited to, biodegradable polymer capsules or bullets, non-degradable, diffusible polymer capsules and biodegradable polymer wafers.
  • According to another embodiment, the invention provides an implantable medical device coated with a compound or a composition comprising a compound of this invention, such that said compound is therapeutically active.
  • According to another embodiment, the invention provides an implantable drug release device impregnated with or containing a compound or a composition comprising a compound of this invention, such that said compound is released from said device and is therapeutically active.
  • Where an organ or tissue is accessible because of removal from the patient, such organ or tissue may be bathed in a medium containing a composition of this invention, a composition of this invention may be painted onto the organ, or a composition of this invention may be applied in any other convenient way.
  • In another embodiment, a composition of this invention further comprises a second therapeutic agent. In one embodiment, the second therapeutic agent is one or more additional compounds of the invention.
  • In another embodiment, the second therapeutic agent may be selected from any compound or therapeutic agent known to have or that demonstrates advantageous properties when administered with a compound having the same mechanism of action as the CRF1 receptor compound of Formula I.
  • In a particular embodiment, the second therapeutic is an agent useful in the treatment or prevention of a disease or condition selected from inflammatory bowel disease (peripherally acting), irritable bowel syndrome (IBS), stress response (colonic motor activity), anxiety, sleep disorder, addictive behavior, acute and chronic neurodegeneration, preterm labor and pain.
  • In another embodiment, the second therapeutic is an agent useful in the treatment or prevention of IBS.
  • In one embodiment, the invention provides separate dosage forms of a compound of this invention and one or more of any of the above-described second therapeutic agents, wherein the compound and second therapeutic agent are associated with one another. The term “associated with one another” as used herein means that the separate dosage forms are packaged together or otherwise attached to one another such that it is readily apparent that the separate dosage forms are intended to be sold and administered together (within less than 24 hours of one another, consecutively or simultaneously).
  • In the pharmaceutical compositions of the invention, the compound of the present invention is present in an effective amount. As used herein, the term “effective amount” refers to an amount which, when administered in a proper dosing regimen, is sufficient to treat (therapeutically or prophylactically) the target disorder. For example, and effective amount is sufficient to reduce or ameliorate the severity, duration or progression of the disorder being treated, prevent the advancement of the disorder being treated, cause the regression of the disorder being treated, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy. Preferably, the compound is present in the composition in an amount of from 0.1 to 50 wt. %, more preferably from 1 to 30 wt. %, most preferably from 5 to 20 wt. %.
  • The interrelationship of dosages for animals and humans (based on milligrams per meter squared of body surface) is described in Freireich et al., (1966) Cancer Chemother. Rep 50: 219. Body surface area may be approximately determined from height and weight of the patient. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardsley, N.Y., 1970, 537.
  • For pharmaceutical compositions that comprise a second therapeutic agent, an effective amount of the second therapeutic agent is between about 20% and 100% of the dosage normally utilized in a monotherapy regime using just that agent. Preferably, an effective amount is between about 70% and 100% of the normal monotherapeutic dose. The normal monotherapeutic dosages of these second therapeutic agents are well known in the art. See, e.g., Wells et al., eds., Pharmacotherapy Handbook, 2nd Edition, Appleton and Lange, Stamford, Conn. (2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif. (2000), each of which references are incorporated herein by reference in their entirety.
  • The compounds for use in the method of the invention can be formulated in unit dosage form. The term “unit dosage form” refers to physically discrete units suitable as unitary dosage for subjects undergoing treatment, with each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, optionally in association with a suitable pharmaceutical carrier. The unit dosage form can be for a single daily treatment dose or one of multiple daily treatment doses (e.g., about 1 to 4 or more times per day). When multiple daily treatment doses are used, the unit dosage form can be the same or different for each dose.
  • Methods of Treatment
  • As used herein the term “subject” and “patient” typically means a human, but can also be an animal in need of treatment, e.g., companion animals (dogs, cats, and the like), farm animals (cows, pigs, horses, sheep, goats, and the like) and laboratory animals (rats, mice, guinea pigs, and the like).
  • The terms “treat” and “treating” are used interchangeably and include both therapeutic treatment and prophylactic treatment (reducing the likelihood of development). Both terms mean decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of a disease (e.g., a disease or disorder delineated herein), lessen the severity of the disease or improve the symptoms associated with the disease.
  • “Disease” means any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
  • As used herein, the term “effective amount” refers to an amount which, when administered in a proper dosing regimen, is sufficient to treat (therapeutically or prophylactically) the target disorder. For example, and effective amount is sufficient to reduce or ameliorate the severity, duration or progression of the disorder being treated, prevent the advancement of the disorder being treated, cause the regression of the disorder being treated, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy.
  • The invention also includes methods of treating diseases, disorders or pathological conditions which benefit from modulation of the CRF1 receptor comprising administering an effective amount of an CRF1 receptor compound of the invention to a subject in need thereof. Diseases and conditions which can benefit from modulation (inhibition or activation) of the CRF1 receptor include, but are not limited to, inflammatory bowel disease (peripherally acting), irritable bowel syndrome (IBS), stress response (colonic motor activity), anxiety, sleep disorder, addictive behavior, acute and chronic neurodegeneration, preterm labor and pain.
  • In one embodiment, an effective amount of a compound of this invention can range from about 0.005 mg to about 5000 mg per treatment. In more specific embodiments, the range is from about 0.05 mg to about 1000 mg, or from about 0.5 mg to about 500 mg, or from about 5 mg to about 50 mg. Treatment can be administered one or more times per day (for example, once per day, twice per day, three times per day, four times per day, five times per day, etc.). When multiple treatments are used, the amount can be the same or different. It is understood that a treatment can be administered every day, every other day, every 2 days, every 3 days, every 4 days, every 5 days, etc. For example, with every other day administration, a treatment dose can be initiated on Monday with a first subsequent treatment administered on Wednesday, a second subsequent treatment administered on Friday, etc. Treatment is typically administered from one to two times daily. Effective doses will also vary, as recognized by those skilled in the art, depending on the diseases treated, the severity of the disease, the route of administration, the sex, age and general health condition of the patient, excipient usage, the possibility of co-usage with other therapeutic treatments such as use of other agents and the judgment of the treating physician.
  • Alternatively, the effective amount of a compound of the invention is from about 0.01 mg/kg/day to about 1000 mg/kg/day, from about 0.1 mg/kg/day to about 100 mg/kg/day, from about 0.5 mg/kg/day to about 50 mg/kg/day, or from about 1 mg/kg/day to 10 mg/kg/day.
  • In another embodiment, any of the above methods of treatment comprises the further step of co-administering to said patient one or more second therapeutic agents. The choice of second therapeutic agent may be made from any second therapeutic agent known to be useful for co-administration with a compound that modulates the CRF1 receptor. The choice of second therapeutic agent is also dependent upon the particular disease or condition to be treated.
  • The term “co-administered” as used herein means that a second therapeutic agent may be administered together with a compound of this invention as part of a single dosage form (such as a composition of this invention comprising a compound of the invention and an second therapeutic agent) or as separate, multiple dosage forms. Alternatively, the additional agent may be administered prior to, consecutively with, or following the administration of a compound of this invention. In such combination therapy treatment, both the compounds of this invention and the second therapeutic agent(s) are administered by conventional methods. The administration of a composition of this invention, comprising both a compound of the invention and a second therapeutic agent, to a subject does not preclude the separate administration of that same therapeutic agent, any other second therapeutic agent or any compound of this invention to said subject at another time during a course of treatment.
  • In one embodiment of the invention, where a second therapeutic agent is administered to a subject, the effective amount of the compound of this invention is less than its effective amount would be where the second therapeutic agent is not administered. In another embodiment, the effective amount of the second therapeutic agent is less than its effective amount would be where the compound of this invention is not administered. In this way, undesired side effects associated with high doses of either agent may be minimized. Other potential advantages (including without limitation improved dosing regimens and/or reduced drug cost) will be apparent to those of skill in the art.
  • Kits
  • The present invention also provides kits for use to treat the target disease, disorder or condition. These kits comprise (a) a pharmaceutical composition comprising a compound of Formula I, or a salt thereof, wherein said pharmaceutical composition is in a container; and (b) instructions describing a method of using the pharmaceutical composition to treat the target disease, disorder or condition.
  • The container may be any vessel or other sealed or sealable apparatus that can hold said pharmaceutical composition. Examples include bottles, ampules, divided or multi-chambered holders bottles, wherein each division or chamber comprises a single dose of said composition, a divided foil packet wherein each division comprises a single dose of said composition, or a dispenser that dispenses single doses of said composition. The container can be in any conventional shape or form as known in the art which is made of a pharmaceutically acceptable material, for example a paper or cardboard box, a glass or plastic bottle or jar, a re-sealable bag (for example, to hold a “refill” of tablets for placement into a different container), or a blister pack with individual doses for pressing out of the pack according to a therapeutic schedule. The container employed can depend on the exact dosage form involved, for example a conventional cardboard box would not generally be used to hold a liquid suspension. It is feasible that more than one container can be used together in a single package to market a single dosage form. For example, tablets may be contained in a bottle, which is in turn contained within a box. In one embodiment, the container is a blister pack.
  • The kits of this invention may also comprise a device to administer or to measure out a unit dose of the pharmaceutical composition. Such device may include an inhaler if said composition is an inhalable composition; a syringe and needle if said composition is an injectable composition; a syringe, spoon, pump, or a vessel with or without volume markings if said composition is an oral liquid composition; or any other measuring or delivery device appropriate to the dosage formulation of the composition present in the kit.
  • In certain embodiment, the kits of this invention may comprise in a separate vessel of container a pharmaceutical composition comprising a second therapeutic agent, such as one of those listed above for use for co-administration with a compound of this invention.
  • General Methods for Preparing CRF1 Receptor Compounds Synthesis of Peptides
  • The peptide component (P) of the compounds of the invention can be synthesized by incorporating orthogonally protected amino acids in a step-wise fashion. Any suitable synthetic methods can be used. Traditional Fmoc or Boc chemistry can be easily adapted to provide the desired peptide component (P) of the compounds of the invention. Fmoc is generally preferred, because the cleavage of the Fmoc protecting group is milder than the acid deprotection required for Boc cleavage, which requires repetitive acidic deprotections that lead to alteration of sensitive residues, and increase acid catalyzed side reactions. (G. B. FIELDS et al. in Int. J. Pept. Protein, 1990, 35, 161).
  • The peptides can be assembled linearly via Solid Phase Peptide Synthesis (SPPS), can be assembled in solution using modular condensations of protected or unprotected peptide components or a combination of both.
  • Solid Phase Peptide Synthesis
  • For SPPS, an appropriate resin is chosen that will afford the desired moiety on the C-terminus upon cleavage. For example upon cleavage of the linear peptide, a Rink amide resin will provide a primary amide on the C-terminus, whereas a Rink acid resin will provide an acid. Rink acid resins are more labile than Rink amide resins and the protected peptide could also be cleaved and subsequently the free acid activated to react with amines or other nucleophiles. Alternatively, other resins could provide attachment of other moieties prior to acylation, leading to cleavage of an alkylated secondary amide, ester or other desired C-terminal modification. A review of commonly used resins and the functional moiety that results after cleavage can be found in manufacturer literature such as NovaBiochem or Advanced Chemtech catalogues.
  • Typically a resin is chosen such that after cleavage the C-terminus is an amide bond. Rink amide resin is a resin that results in a C-terminal amide during cleavage. The orthogonally protected Fmoc amino acids are added stepwise using methods well known in literature (Bodansky M. Principles of Peptide synthesis (1993) 318p; Peptide Chemistry, a Practical Textbook (1993); Spinger-Verlag). These procedures could be done manually or by using automated peptide synthesizers.
  • The process involves activating the acid moiety of a protected amino acid, using activating agents such as HBTU, HATU, PyBop or simple carbodiimides. Often an additive is used to decrease racemization during coupling such as HOBt or HOAt (M. SCHNÖLZER et al., Int. J. Pept. Protein Res., 1992, 40, 180). Manually, the coupling efficiency can be determined photometrically using a ninhydrin assay. If the coupling efficiency is below 98%, a second coupling may be desired. After the second coupling a capping step may be employed to prevent long deletion sequences to form, simplifying the purification of the desired final compound. With automation, second couplings are not commonly required, unless a residue is known to be problematic such as Arginine.
  • Deprotection of the Fmoc is most commonly accomplished using piperidine (20%) in dimethylformamide (DMF). Alternatively other secondary amines may also be used such as morpholine, diethylamine or piperazine. This reaction is facile and normally is accomplished within 20 minutes using piperidine. After deprotection the resin is washed several times with DMF and DCM prior to coupling with the next residue. This process is repeated, assembling the peptide linearly until the sequence is complete. The final Fmoc is removed, which allows for coupling with the tether moiety.
  • In a preferred synthesis, the peptide is formed by SPPS accomplished manually or in an automated fashion using a commercially available synthesizer such as the CEM Microwave peptide synthesizer, Rainin Symphony synthesizer, or ABI 433 flow-through synthesizer. Commercially available Rink Amide resin is used for synthesizing the C-terminal amide peptides (Rink, H. Tetrahedron Lett, 28, 4645, 1967). Peptide synthesis reagents (coupling, deprotection agents) are commercially available and include HOBT, HBTU (Novabiochem) as well as DMF, DCM, Piperidine, NMP, and DIEA (Sigma-Aldrich). Suitably protected amino acids for use in solid phase peptide synthesis are commercially available from many sources, including Sigma-Aldrich and CEM Corporation.
  • For example, a convenient preparation of peptides on a 0.1 mmol or 0.25 mmol scale uses Rink amide solid-phase resin with a substitution of about 0.6 mmol/g. Linear attachment of the amino acids is accomplished on a ABI continuous flow automated synthesizer using 5 eq of orthogonally protected amino acid (AA), and using HBTU/HOBt coupling protocol, (5 eq. of each reagent). In another preferred synthesis, peptides can be synthesized using a microwave instrument using 10 eq of reagents. Deprotection of Fmoc can be accomplished with 20% piperidine in DMF followed by washing with DMF and DCM.
  • In both cases (i.e., Rink acid and Rink amide resins), final Fmoc deprotection of the N-terminus would leave a free amine after cleavage from the resin unless it is modified prior to cleavage. In the compounds of the invention, tether moieties are attached through amide bonds.
  • Solution Phase Synthesis of Peptides
  • For solution phase synthesis the desired peptide is generally broken down into peptide fragments in units of 2-4 amino acids. The selected unit is dependent on the sequence, the stability of the fragment to racemization, and the ease of assembly. As each amino acid is added, only 1-1.5 eq of the residue is required, versus the 5-10 equivalents of reagent required for SSPS. Preactivated amino acids such as OSu active ester and acid fluorides also can be used, requiring only a base for completion of the reaction.
  • Coupling times require 1.5-2 hours for each step. Two fragments are condensed in solution, giving a larger fragment that then can be further condensed with additional fragments until the desired sequence is complete. The solution phase protocol uses only 1 eq of each fragment and will use coupling reagents such as carbodiimides (DIC). For racemized prone fragments, PyBop or HBTU/HOBt can be used. Amino acids with Bsmoc/tBu or Fmoc/tBu and Boc/Benzyl protection are equally suitable for use.
  • When Fmoc is used, the use of 4-(aminomethyl)piperidine or tris(2-aminoethyl)amine as the deblocking agent can avoid undesired side reactions. The resulting Fmoc adduct can be extracted with a phosphate aqueous buffer of pH 5.5 (Organic Process Research & Development 2003, 7, 2837). If Bsmoc is used, no buffer is required, only aqueous extractions are needed. Deprotections using these reagents occur in 30-60 minutes. Deblocking of the Fmoc group on the N-terminal residue provides a free terminal amine that is used for attachment of the tether moiety. In the compounds of the invention, tether moieties are attached through amide bonds to the N-terminal amine.
  • One advantage of solution phase synthesis is the ability to monitor the compound after every coupling step by mass spectrometry to see that the product is forming. In addition, a simple TLC system could be used to determine completion of reaction.
  • Attachment of Tethers
  • Tethers are attached to the terminal nitrogen of the N-terminal amino acid of the peptide chain using amide bond coupling:
  • Figure US20110301087A1-20111208-C00062
  • The tether can be attached using solid phase procedures or in solution using an amide bond coupling. After the N-terminus is suitably coupled, the final compound is cleaved from the resin using an acidic cocktail (Peptide Synthesis and Applications, John Howl, Humana Press, 262p, 2005). Typically these cocktails use concentrated trifluoroacetic acid (80-95%) and various scavengers to trap carbocations and prevent side chain reactions. Typical scavengers include isopropylsilanes, thiols, phenols and water. The cocktail mixture is determined by the residues of the peptide. Special care needs to be taken with sensitive residues, such as methionine, aspartic acid, and cysteine. Typical deprotection occurs over 2-5 hours in the cocktail. A preferred deprotection cocktail include the use of triisopropylsilane (TIS), Phenol, thioanisole, dodecanethiol (DDT) and water. Methane sulfonic acid (MSA) may also be used in the cocktail (4.8%). A more preferred cocktail consists of (TFA:MSA:TIS:DDT:Water 82:4.5:4.5:4.5:4.5; 10 mL/0.1 mmol resin).
  • After deprotection, the resin is removed via filtration, and the final compound is isolated via precipitation from an organic solvent such as diethyl ether, m-tert-butyl ether, or ethyl acetate and the resulting solid collected via filtration or lyophilized to a powder. Purification of the peptide using reverse phase HPLC may be required to achieve sufficient purity. Generally, a gradient of aqueous solvent with an organic solvent will provide sufficient separation from impurities and deletion sequences. Typically 0.1% TFA is used as the aqueous and organic modifier, however, other modifiers such as ammonium acetate can also be used. After purification, the compound is collected, analyzed and fractions of sufficient purity are combined and lyophilized, providing the compound as a solid.
  • Amino Acid Reagents
  • The following commercially available orthogonally protected amino acids used can be used in the synthesis of compounds of the invention: Fmoc-Tyr(tBu)-OH, Fmoc-Ala-OH*H2O, Fmoc-Arg(Pbf)-OH, Fmoc, Asn(Trt)-OH, Fmoc-Asp(tBu), Fmoc-Cys(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glx(Pbf)-OH, Fmoc-Gly-OH, Fmoc-His(Trt)-OH, Fmoc-Leu-OH, Fmoc-Ile-OH, Fmoc, Lys(tBu)-OH, Fmoc-Met-OH, Fmoc-Phe-OH, Fmoc-Ser(tBu)-OH, Fmoc-Thr(tBu)-OH, Fmoc-Typ-OH, and Fmoc-Val-OH. Additional amino acids suitable for incorporation into the compounds of the invention (e.g., D amino acids, substituted amino acids and other protecting group variations) are also commercially available or synthesized by methods known in the art.
  • Analytical Methods
  • The compounds of the invention are analyzed for purity by HPLC using the methods listed below. Purification is achieved by preparative HPLC.
  • Fast LC/MS Method
  • Column: Phenomenex Luna C-5 20×30 mm
  • Flow: 1.0 ml/min
  • Solvent A: 0.1% TFA in Type I water
  • Solvent B: 0.1% TFA in Acetonitrile
  • UV 220 nm
  • Injection: 20 ul
  • Gradient 5-95% B (7 minutes); 95-5% B (1 minute); 5% B (4 minutes)
  • Analytical Purity Method
  • Column: Phenomenex Luna C-5 20×30 mm
  • Flow: 1.0 ml/min
  • Solvent A: 0.1% TFA in Type I water
  • Solvent B: 0.1% TFA in Acetonitrile
  • UV: 220 nm
  • Injection: 20 ul
  • Gradient: 2-95% B (10 minutes); 95-2% B (2 minutes); 2% B (2 minutes)
  • Preparative LC/MS Method
      • Column: Phenomenex Luna C-5 250×150 mm
      • Flow: 5.0 ml/min
      • Solvent A: 0.1% TFA in Type I water
      • Solvent B: 0.1% TFA in Acetonitrile
      • UV: 220 nm
      • Injection: 900 ul
      • Gradient: 35% B (5 minutes); 35-85% B (13 minutes); 85-35% B (0.5 minutes); 35% B (1.5 minutes)
    Synthesis of Selected Compounds
  • Compound 58 Pal-RILMTKLRASTTS-amide
  • Compound 58 was synthesized as described above on Rink amide resin at 0.1 mmol scale. Amino acids were coupled sequentially as described above. Following deprotection of the Fmoc group on the N-terminal residue serine, the N-terminal amine was capped with palmitic acid (10 eq.), HBTU (10 eq.) and DIEA (10 eq.) as described above. The pepducin was cleaved from the resin by TFA containing MS, TIS, DDT, and water (82:4.5:4.5:4.5:4.5; mL), filtered through a Medium frit Buchner full, triturated with ether and the resulting precipitate collected by centrifugation. Crude peptide was taken up in minimum amount of DMSO and purified by RP-HPLC as described previously. Fractions with correct MW were pooled and lyophilized and analyzed for purity using Method A. The yield of representative lots is illustrated in the following table.
  • Lot # Yield (mg)
    1 3.3
    2 5
  • Compound 32 Pal-TDRLRKWMFI-amide

    Compound 32 was synthesized as described for Compound 58. The yield of representative lots is illustrated in the following table.
  • Lot # Yield (mg)
    1 6
  • Compound 29 Pal-GSTDRLRKWMFI-amide

    Compound 29 was synthesized as described for Compound 58. The yield of representative lots is illustrated in the following table.
  • Lot # Yield (mg)
    1 4
    2 2.1
    3 2.6
    4 12.3

    The compounds listed in Tables 6-9 were prepared according to the methods described herein.
  • TABLE 6A
    CRF1 i1 loop pepducins
    Compound Sequence N terminus C terminus MS Theoretical MS Observed
    Compound 1 RSLRNIIH C15H31C(O)— Amide 623.8015 623.6
    Compound 2 RSIRSLRNIIH C15H31C(O)— Amide 802.0115 801.6
    Compound 3 GSGRLRSIRSLRNIIH C15H31C(O)— Amide 691.849 692
    Compound 4 LFIRIRSIRSLRNIIH C15H31C(O)— Amide 1123.428 1122.9
    Compound 5 RLRSIRSLRNIIHWN C15H31C(O)— Amide 724.893 725
    Compound 6 RLRSIRSLRNIIH C15H31C(O)— Amide 624.789 624.85
    Compound 7 RLRSIRSLRN C15H31C(O)— Amide 754.956 755
    Compound 8 RLRSIRSLR C15H31C(O)— Amide 697.905 697.7
    Compound 9 RSIRSLRN C15H31C(O)— Amide 620.2845 620
    Compound 10 GSGRLRSIRSLRNIIHWN C15H31C(O)— Amide 1187.4295 1187.4
    Compound 11 RSIRSLRNIIHWN C15H31C(O)— Amide 952.168 951.8
    Compound 13 RSLRNIIHWN C15H31C(O)— Amide 773.9575
  • TABLE 6B
    CRF1 i1 loop pepducin structures
    Compound Number and Structure
    Compound 1
    Figure US20110301087A1-20111208-C00063
    Compound 2
    Figure US20110301087A1-20111208-C00064
    Compound 3
    Figure US20110301087A1-20111208-C00065
    Compound 4
    Figure US20110301087A1-20111208-C00066
    Compound 5
    Figure US20110301087A1-20111208-C00067
    Compound 6
    Figure US20110301087A1-20111208-C00068
    Compound 7
    Figure US20110301087A1-20111208-C00069
    Compound 8
    Figure US20110301087A1-20111208-C00070
    Compound 9
    Figure US20110301087A1-20111208-C00071
    Compound 10
    Figure US20110301087A1-20111208-C00072
    Compound 11
    Figure US20110301087A1-20111208-C00073
    Compound 13
    Figure US20110301087A1-20111208-C00074
  • TABLE 7A
    CRF1 i2 loop pepducins
    C MS MS
    Compd # Sequence N terminus terminus Theoretical Observed
    Compound 14 TYSTDRLRK C15H31C(O)— Amide 689.343 689.45
    Compound 15 TYSTDRLRKWMFI C15H31C(O)— Amide 978.212 977.95
    Compound 16 GSTDRLRK C15H31C(O)— Amide 585.73 585.8
    Compound 17 GSTDRLRKMFI C15H31C(O)— Amide 781.494 781
    Compound 18 GSGTYSTDRLRK C15H31C(O)— Amide 789.933 789.5
    Compound 19 STDRLRKMF C15H31C(O)— Amide 696.3895 696
    Compound 20 STDRLRKWMF C15H31C(O)— Amide 789.4945 789
    Compound 21 GSGTYSTDRLRKWMF C15H31C(O)— Amide 1022.223 1021.5
    Compound 22 GSTDRLRRWMFI C15H31C(O)— Amide 888.6055 888.1
    Compound 23 GSGTYSTDRLRKWMFI C15H31C(O)— Amide 1078.71 1078.4
    Compound 24 GSGRLRKWM C15H31C(O)— Amide 664.8635 664.5
    Compound 25 GSGTYDRLRKWM C15H31C(O)— Amide 853.985 854
    Compound 26 GSGRLRKWMFI C15H31C(O)— Amide 795.029 794.6
    Compound 27 GRLRKWMFI C15H31C(O)— Amide 722.9645 722.7
    Compound 28 GSRLRKWMFI C15H31C(O)— Amide 766.5035 766.2
    Compound 29 GSTDRLRKWMFI C15H31C(O)— Amide 874.599 875
    Compound 30 RLRKWMFI C15H31C(O)— Amide 694.439 694.2
    Compound 31 DRLRKWMFI C15H31C(O)— Amide 751.9825 751.7
    Compound 32 TDRLRKWMFI C15H31C(O)— Amide 802.5345 802.2
    Compound 33 STDRLRKWMFI C15H31C(O)— Amide 846.0735 846.1
    Compound 34 YSTDRLRKWMFI C15H31C(O)— Amide 927.66 927.3
    Compound 35 LTYSTDRLRKWMFI C15H31C(O)— Amide 690.1936667 690.7
    Compound 36 VLTYSTDRLRKWMFI C15H31C(O)— Amide 1084.356 1083.7
    Compound 37 GSTDRLRKWMFA C15H31C(O)— Amide 853.559 853.2
    Compound 38 GSTDRLRKWMAI C15H31C(O)— Amide 836.551 836
    Compound 39 GSTDRLRKWAFI C15H31C(O)— Amide 844.54 844.5
    Compound 40 GSTDRLRKAMFI C15H31C(O)— Amide 817.033 816.6
    Compound 41 RLRKWMAI C15H31C(O)— Amide 656.391 656
    Compound 42 DRLRKWMAI C15H31C(O)— Amide 713.935 713.5
    Compound 43 TDRLRKWMAI C15H31C(O)— Amide 764.4865 764.2
    Compound 44 ADRLRKWMFI C15H31C(O)— Amide 787.5215 786.8
    Compound 45 TARLRKWMFI C15H31C(O)— Amide 780.53 780.2
    Compound 46 TDALRKWMFI C15H31C(O)— Amide 759.981 759.2
    Compound 47 TDRARKWMFI C15H31C(O)— Amide 781.495 780.7
    Compound 48 TDRLAKWMFI C15H31C(O)— Amide 759.981 759.1
    Compound 49 TDRLRAWMFI C15H31C(O)— Amide 773.9875 773.2
    Compound 50 TDRLRKAMFI C15H31C(O)— Amide 744.9685 744.3
    Compound 51 TDRLRKWAFI C15H31C(O)— Amide 772.4755 771.8
    Compound 52 TDRLRKWMFA C15H31C(O)— Amide 781.495 780.8
    Compound 53 GSTDRLRKAXFI C15H31C(O)— Amide 808.0135 807.3
    Compound 54 TDRLRKWXFI C15H31C(O)— Amide 793.5155 792.8
  • TABLE 7B
    CRF1 i2 loop pepducin structures
    Compound Number and Structure
    Compound 14
    Figure US20110301087A1-20111208-C00075
    Compound 15
    Figure US20110301087A1-20111208-C00076
    Compound 16
    Figure US20110301087A1-20111208-C00077
    Compound 17
    Figure US20110301087A1-20111208-C00078
    Compound 18
    Figure US20110301087A1-20111208-C00079
    Compound 19
    Figure US20110301087A1-20111208-C00080
    Compound 20
    Figure US20110301087A1-20111208-C00081
    Compound 21
    Figure US20110301087A1-20111208-C00082
    Compound 22
    Figure US20110301087A1-20111208-C00083
    Compound 23
    Figure US20110301087A1-20111208-C00084
    Compound 24
    Figure US20110301087A1-20111208-C00085
    Compound 25
    Figure US20110301087A1-20111208-C00086
    Compound 26
    Figure US20110301087A1-20111208-C00087
    Compound 27
    Figure US20110301087A1-20111208-C00088
    Compound 28
    Figure US20110301087A1-20111208-C00089
    Compound 29
    Figure US20110301087A1-20111208-C00090
    Compound 30
    Figure US20110301087A1-20111208-C00091
    Compound 31
    Figure US20110301087A1-20111208-C00092
    Compound 32
    Figure US20110301087A1-20111208-C00093
    Compound 33
    Figure US20110301087A1-20111208-C00094
    Compound 34
    Figure US20110301087A1-20111208-C00095
    Compound 35
    Figure US20110301087A1-20111208-C00096
    Compound 36
    Figure US20110301087A1-20111208-C00097
    Compound 37
    Figure US20110301087A1-20111208-C00098
    Compound 38
    Figure US20110301087A1-20111208-C00099
    Compound 39
    Figure US20110301087A1-20111208-C00100
    Compound 40
    Figure US20110301087A1-20111208-C00101
    Compound 41
    Figure US20110301087A1-20111208-C00102
    Compound 42
    Figure US20110301087A1-20111208-C00103
    Compound 43
    Figure US20110301087A1-20111208-C00104
    Compound 44
    Figure US20110301087A1-20111208-C00105
    Compound 45
    Figure US20110301087A1-20111208-C00106
    Compound 46
    Figure US20110301087A1-20111208-C00107
    Compound 47
    Figure US20110301087A1-20111208-C00108
    Compound 48
    Figure US20110301087A1-20111208-C00109
    Compound 49
    Figure US20110301087A1-20111208-C00110
    Compound 50
    Figure US20110301087A1-20111208-C00111
    Compound 51
    Figure US20110301087A1-20111208-C00112
    Compound 52
    Figure US20110301087A1-20111208-C00113
    Compound 53
    Figure US20110301087A1-20111208-C00114
    Compound 54
    Figure US20110301087A1-20111208-C00115
  • TABLE 8A
    CRF1 i3 loop pepducins
    Compound Sequence N terminus C terminus MS Theoretical MS Observed
    Compound 55 SETIQYRKAVKA C15H31C(O)— Amide 816.506 816.1
    Compound 56 TIQYRKAVKA C15H31C(O)— Amide 708.41 708.2
    Compound 57 SETIQYRKAVKATL C15H31C(O)— Amide 923.6365 923.7
    Compound 58 RILMTKLRASTTS C15H31C(O)— Amide 858.598 858
    Compound 59 RILMTKLRA C15H31C(O)— Amide 670.417 670
    Compound 60 ASTTSETIQYR C15H31C(O)— Amide 747.8715 747.4
    Compound 61 ASTTSETIQYRKAVKA C15H31C(O)— Amide 664.7916667 664.85
    Compound 62 ASTTSETIQYRKAVKS C15H31C(O)— Amide 670.1246667 670.2
  • TABLE 8B
    CRF1 i3 loop pepducin structures
    Compound Number and Structure
    Compound 55
    Figure US20110301087A1-20111208-C00116
    Compound 56
    Figure US20110301087A1-20111208-C00117
    Compound 57
    Figure US20110301087A1-20111208-C00118
    Compound 58
    Figure US20110301087A1-20111208-C00119
    Compound 59
    Figure US20110301087A1-20111208-C00120
    Compound 60
    Figure US20110301087A1-20111208-C00121
    Compound 61
    Figure US20110301087A1-20111208-C00122
    Compound 62
    Figure US20110301087A1-20111208-C00123
  • TABLE 9A
    CRF1 i4 loop pepducins
    Compound Sequence N terminus C terminus MS Theoretical MS Observed
    Compound 63 VRSAIRKRWHRW C15H31C(O)— Amide 945.1785 945.1
    Compound 64 GSIRARVARAM C15H31C(O)— Amide
  • TABLE 9B
    CRF1 i4 loop pepducin structures
    Compound Number and Structure
    Compound 63
    Figure US20110301087A1-20111208-C00124
    Compound 64
    Figure US20110301087A1-20111208-C00125
  • Methods of Screening Functional Assays
  • Functional assays suitable for use in detecting and characterizing GPCR signaling include Gene Reporter Assays and Calcium Flux assays, cAMP and kinase activation assays. Several suitable assays are described in detail below.
  • Gene Reporter Assays
  • Cells expressing the CRF1 receptor can be transiently or stably transfected with a reporter gene plasmid construct containing an enhancer element which responds to activation of a second messenger signaling pathway or pathways, thereby controlling transcription of a cDNA encoding a detectable reporter protein. CRF1 expression can be the result of endogenous expression on a cell line or cell type or the result of stable or transient transfection of DNA encoding the receptor of interest into a cell line by means commonly used in the art. Immortalized cell lines or primary cell cultures can be used. For example, HEK293 cells can be co-transfected with expression plasmids encoding the GPCR CRFR1 and a reporter plasmid containing firefly luciferse cDNA whose transcription is regulated by tandem repeats of a cAMP response element (CRE-luc).
  • To test for agonist or inverse agonist activity, cells containing the GPCR and the reporter gene construct can be challenged by the test compound for a predetermined period of time (e.g., 2-12 hours, typically 4 hours). Cells can then be assessed for levels of reporter gene product. If the activated pathway is stimulatory (e.g., Gs or Gq), agonist activity results in activation of transcription factors, in turn causing an increase in reporter gene transcription, detectable by an increase in reporter activity. Inverse agonists will suppress levels of reporter to below basal levels in a dose dependent manner. For example, cells transfected with CRF1 and CRE-luc are challenged by test compounds. Agonist response through Gs activation will cause an increase in cAMP levels in the cells containing both components of the system. Increased cAMP levels result in an increased rate of transcription of the CRE-regulated luciferase cDNA. This increased transcription results in an increased level of active luciferase enzyme, detectable as an increase in luminescence relative to unstimulated cells.
  • To test for antagonist or inhibitory activity through a stimulatory pathway, cells containing both the GPCR and the reporter gene construct can be activated by a receptor agonist to increase gene reporter product levels. Treatment with antagonists will counter the effect of agonist stimulation in a dose- and receptor-dependent manner. To test for agonist activity on receptor signaling through an inhibitory pathway (eg, Gi), cells can be treated with a systematic activator (e.g., forskolin) to increase levels of reporter gene product. Activation of Gi by treatment with receptor agonist will inhibit this expression by inhibiting adenylyl cyclase.
  • To screen for antagonist activity, test compounds can be assessed for the ability to counter agonist inhibition of adenylyl cyclase, resulting in increase reporter transcription.
  • Alternatively, a plasmid construct expressing the promiscuous G-protein Gal 6 can be used to obtain a positive signal from a GPCR which normally couples to an inhibitory G-protein. Co-expression of the chimeric G-protein Gaq/Gai5 (Coward et al. Analytical Biochemistry 270, 242-248 (1999)) allows coupling to Gi-coupled receptors and conversion of second messenger signaling from the inhibitory Gi pathway to the stimulatory Gq pathway. Agonist and antagonist assessment in these systems is the same as the stimulatory pathways. Well-to-well variation caused by such factors as transfection efficiency, unequal plating of cells, and cell survival rates can be normalized in transient transfection assays by co-transfecting a constitutively expressing reporter gene with a non-interfering signal independent of the regulated reporter.
  • Gene Reporter Assay for the CRF1
  • CRF1 signals predominantly through Gs. HEK293 cells stably expressing CRF1 can be seeded in a white opaque 96-well tissue culture plate (CulturePlate, Perkin Elmer) and can be transiently transfected with CRE (cAMP Response Element)-luciferase (Stratagene PathDetect) (1 ug/ml) using LipofectAMINE (Invitrogen) according to manufacturer's protocol in serum-free DMEM. 16-24 hours post-transfection, media can be replaced with DMEM supplemented with 0.1-0.2% BSA with or without ligand and test compound for 4 hours. The media can be removed and cells can be lysed with 50 ul SteadyLite HTS (Perkin Elmer) reconstituted in Substrate Buffer and diluted 1:2 in PBS. After 10 minutes, luminescence can be measured using a TopCount NXT microplate reader.
  • When screening for antagonists, optimal agonist (CRF or urocortin I) concentrations can be determined by experimentally derived calculations of EC50 values. Activity linked to Gq signaling can be assessed using the above procedure substituting SRE-luciferase or NFAT-luciferase as the reporter gene.
  • Calcium Flux Assay
  • Calcium Flux Assay is one of the most widely used cell-based GPCR functional assays. It most often uses calcium sensing fluorescent dyes such as fura2 AM, fluo-4 and Calcium-4 to measure changes in intracellular calcium concentration. It is used mainly to detect GPCR signaling via Gaq subunit, but can be used to measure signaling through any pathway which leads to a detectable increase in intracellular calcium. Activation of these Gq-coupled GPCRs leads to activation of phospholipase C, which subsequently leads to increase in inositol phosphate production. IP3 receptors on endoplasmic reticulum sense the change then release calcium into cytoplasm. Intracellular calcium binding to the fluorescent dyes can be detected by instruments that quantify fluorescent intensities, such as FLIPR Tetra, Flexstation (MDS) and FDSS (Hamamatsu). In additional to assess Gq-couple receptor signaling, calcium flux assay can also be used to study Gs and Gi couple receptors by co-expressing CNG (cycic nucleotide gated calcium channel) or chimeric G-proteins (Gqi5, Gsi5 for example). Activation of some Gi-coupled receptors can also be detected by calcium flux assay via Gβγ mediated phospholipase C activation.
  • CRF1 Assessment of Calcium Flux
  • An example of the use of the calcium flux assay can be assessing a-CRF1 activation of CRF1 in Y-79, a human retinoblastoma cell line which endogenously expresses functional CRF1. Y-79 cells can be seeded into 96-well black plates with clear bottom at 200K/well in Hank's balanced salt solution with 20 mM HEPES, 0.1% BSA. After dye loaded by incubating in Calcium-4 dye at room temperature for 1 hour, cell plates can be placed in Flexstation 3. The addition of test compound or reference antagonists can be done either by manual pipetting or by liquid handling on Flexstation. The latter allows the assessment of agonist activity of the test compound. After incubation of 15 minutes at 37° C., CRF can be added on Flexstation and receptor activation can be assessed by measuring changes in fluorescent intensity.
  • HTRF cAMP Assay and IP-One Assay (Cisbio)
  • HTRF (homogeneous time resolved fluorescence) is a technology developed by Cisbio Bioassays based on TR-FRET (time-resolved fluorescence resonance energy transfer). Cisbio Bioassays has developed a wide selection of HTRF-based assays compatible with whole cells, thereby enabling functional assays run under more physiological conditions. cAMP kits are based on a competitive immunoassay using cryptate-labeled anti-cAMP antibody and d2-labeled cAMP. This assay allows the measurement of increase in intracellular cAMP upon Gs-coupled receptor activation as well as decrease in forskolin stimulated increase in cAMP upon Gi-coupled receptor activation. Cells expressing CRF1 endogenously or via transfection can be used to assess compound pharmacology using cAMP assays as a means to measure the activity of adenylyl cyclase resulting from CRF1 activation or inhibition.
  • The IP-One assays are competitive immunoassays that use cryptate-labeled anti-IP1 monoclonal antibody and d2-labeled IP1. IP1 is a relatively stable downstream metabolite of IP3, and accumulates in cells following Gq receptor activation.
  • CRF1 Assessment of cAMP
  • An example of the use of the cAMP assay can be assessing CRF activation of CRF1 in Y-79, a human retinoblastoma cell line which endogenously expresses functional CRF1. Y-79 cells were seeded into 96-well half area black plates at 30K/well in Hank's balanced salt solution with 50 mM HEPES, 0.1% BSA, 200 uM IBMX. Compounds to be tested for agonist activity were added to cells, incubated for 30 minutes at 37 degrees C. and assayed for cAMP concentration. To test for antagonist activity, compounds to be tested were added to cells, followed by the addition of agonist at an experimentally determined optimal concentration, generally the EC80. Plates were incubated for 30 minutes at 37 degrees C. and assayed for cAMP. To determine concentration of cAMP in each well, d2-labeled cAMP in Conjugate/Lysis buffer was added to wells. This addition lysed the cells and input a known concentration of labeled cAMP. This was followed by the addition of anti-cAMP antibody conjugated to cryptate. FRET occurs when cryptate-labeled antibody binds to d2-labeled cAMP. Therefore, the specific signal is inversely proportional to the concentration of cAMP in the sample. Conversion of signal to concentration of cAMP in the well was accomplished by comparing the FRET signal of sample to a standard curve generated using known concentrations of cAMP.
  • Testing of compounds was conducted in this way and the results are set forth in Table 10. For the data in Table 5 IC50 values from: 1 nM to 500 nM are designated as *****; 501 nM to 1000 nM are designated as ****; 1001 nM to 5000 nM are designated as ***; 5001 nM to 10,000 nM are designated as **; and greater than 10,000 nM are designated *.
  • TABLE 10
    CRF1 cAMP screening data (Y79 cells, 300 nM pepducin
    against agonist dose response)
    %
    Inhibition
    Sample Lot Loop Sequence N terminus C terminus IC50 (± SD)
    Compound 58 i3 RILMTKLRASTTS Lipid Amide **** 35
    Compound 62 i3 ASTTSETIQYRKAVKS Lipid Amide *** 50
    Compound 5 it RLRSIRSLRNIIHWN Lipid Amide *** 54 ± 13
    Compound 22 i2 GSTDRLRRWMFI Lipid Amide *** 50 ± 15
    Compound 26 i2 GSGRLRKWMFI Lipid Amide *** 63
    Compound 64 i1 RSIRSLRNIIHWN Lipid Amide *** 55 ± 13
    Compound 28 i2 GSRLRKWMFI Lipid Amide *** 52 ± 5 
    Compound 29 i2 GSTDRLRKWMFI Lipid Amide **** 53 ± 14
    Compound 30 i2 RLRKWMFI Lipid Amide *** 39 ± 7 
    Compound 31 i2 DRLRKWMFI Lipid Amide *** 30 ± 17
    Compound 32 i2 TDRLRKWMFI Lipid Amide **** 53 ± 9 
    Compound 33 i2 STDRLRKWMFI Lipid Amide *** 36 ± 9 
    Compound 34 i2 YSTDRLRKWMFI Lipid Amide **** 21 ± 5 
    Compound 35 i2 LTYSTDRLRKWMFI Lipid Amide *** 26 ± 15
    Compound 36 i2 VLTYSTDRLRKWMFI Lipid Amide * 19 ± 10
    Compound 37 i2 GSTDRLRKWMFA Lipid Amide *** 45 ± 19
    Compound 38 i2 GSTDRLRKWMAI Lipid Amide *** 60 ± 8 
    Compound 40 i2 GSTDRLRKAMFI Lipid Amide **** 61 ± 20
    Compound 41 i2 RLRKWMAI Lipid Amide **** 40
    Compound 42 i2 DRLRKWMAI Lipid Amide ** 65
    Compound 43 i2 TDRLRKWMAI Lipid Amide * 48
    Compound 44 i2 ADRLRKWMFI Lipid Amide * 42
    Compound 45 i2 TARLRKWMFI Lipid Amide * 70
    Compound 46 i2 TDALRKWMFI Lipid Amide * 58
    Compound 47 i2 TDRARKWMFI Lipid Amide *** 62
    Compound 48 i2 TDRLAKWMFI Lipid Amide * 30
    Compound 49 i2 TDRLRAWMFI Lipid Amide * 42
    Compound 50 i2 TDRLRKAMFI Lipid Amide * 58
    Compound 51 i2 TDRLRKWAFI Lipid Amide **** 47 ± 7 
    Compound 52 i2 TDRLRKWMFA Lipid Amide * 70
    Compound 53 i2 GSTDRLRKAXFI Lipid Amide **** 50 ± 14
    Compound 54 i2 TDRLRKWXFI Lipid Amide *** 55 ± 7 

    Receptor Binding assays
  • Peptide ligands can be radio-labeled with 1-125. Test compounds can be assessed by adding increasing amounts of unlabeled test against known tracer amounts of labeled ligand. The test compounds can be assessed for the ability to compete for receptor binding sites (competitive) or to affect the affinity of ligands indirectly (non-competitive). In addition, the affinity of a compound for the receptor can be assessed by the ability of unlabeled compound to displace trace amounts of labeled test compound. Radio-labeled ligands can also be used to assess receptor numbers on cells or membranes.
  • In Vivo Assays
  • CRF1 activation leads to an increase in colonic motility. Peripheral administration of CRF or urocortin I intravenously or intraperitoneally induces propulsive colonic motor events, reduction of colonic transit time, and diarrhea in rodents.
  • Colonic Function
  • Distal colonic function can be measured by assessing the time required to expel a plastic bead inserted into the distal colon of a rat. Efficacy of CRF1 compounds of this invention in this model would be assessed by the ability to reduce colonic motility, thereby lengthening the time to expulsion of the bead.
  • Visceral Pain Associated with Colonic Distension.
  • Colorectal distention results in contractions of the muscles of the abdomin and hind limbs. This response has been used as a quantitative indicator of visceral pain. Visceromotor response can be quantified using the abdominal withdrawal reflex (al-Chaer et al. Gastroenterology 119: 1276-1285 (2000)). Distension is achieved by the insertion into the descending colon and inflation of a latex balloon. A five-part scoring scale relates reflexive actions to intensity of nociception. A pain threshold can be determined by relating pressure in the distension balloon to the onset of a defined pain reflex. Effects of CRFR1 activation on visceral pain due to colonic distension can be assessed by administration of CRF or CRF1-specific agonists. CRF1 receptor compounds of the invention which inhibit CRF1 activation would be expected to increase the pain threshold.
  • The teachings of all patents, published applications and references cited herein are incorporated by reference in their entirety.
  • While this invention has been particularly shown and described with references to example embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims (119)

1. A compound represented by Formula I:

T-L-P,
or pharmaceutically acceptable salts thereof, wherein:
P is a peptide comprising at least three contiguous amino-acid residues of an intracellular i1, i2, i3 loop or an intracellular i4 domain of the CRF1 receptor;
L is a linking moiety represented by C(O) and bonded to P at an N terminal nitrogen of an N-terminal amino-acid residue;
and T is a lipophilic tether moiety bonded to L, wherein the C-terminal amino acid residue of P is optionally functionalized.
2. The compound of claim 1, wherein P comprises at least six contiguous amino acid residues.
3. The compound of claim 1, wherein P is derived from the i1 loop and is represented by the following structural formula or pharmaceutically acceptable salts thereof
X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11-X12-X13-X14-X15-X16- X17-X18-X19-R1
wherein:
X1 is absent or a valine residue;
X2 is absent or a leucine or glycine residue;
X3 is absent or a phenylalanine or serine residue;
X4 is absent or a leucine or glycine residue;
X5 is absent or an arginine residue;
X6 is absent or a leucine or isoleucine residue;
X7 is absent or an arginine residue;
X8 is absent or a serine residue;
X9 is absent or an isoleucine residue;
X10 is absent or an arginine residue;
X11 is absent or a cysteine or serine residue;
X12 is absent or a leucine residue;
X13 is absent or an arginine residue;
X14 is absent or an asparagine residue;
X15 is absent or an isoleucine residue;
X16 is absent or an isoleucine residue;
X17 is absent or a histidine residue;
X18 is absent or tryptophan residue;
X19 is absent or an asparagine residue; provided that at least five of X1-X19 are present;
R1 is OR2 or N(R2)2;
each R2 is independently hydrogen or (C1-C10)alkyl; and
from 0 to 5 amino acid residues are present in the D configuration.
4. The compound of claim 3, wherein at least four of X7, X8, X9, X10, X11, X12, X13 and X14 are present.
5. The compound of claim 4, wherein
X7 is an arginine residue;
X8 is a serine residue;
X9 is an isoleucine residue; and
X10 is an arginine residue.
6. The compound of claim 4, wherein
X11 is a cysteine or serine residue;
X12 is a leucine residue;
X13 is an arginine residue; and
X14 is an asparagine residue.
7. The compound of claim 6, wherein X11 is a cysteine residue.
8. The compound of claim 6, wherein X11 is a serine residue.
9. The compound of claim 3, wherein
X14 is an asparagine residue;
X15 is an isoleucine residue;
X16 is an isoleucine residue; and
X17 is a histidine residue.
10. The compound of claim 3, selected from:
Figure US20110301087A1-20111208-C00126
Figure US20110301087A1-20111208-C00127
Figure US20110301087A1-20111208-C00128
Figure US20110301087A1-20111208-C00129
Figure US20110301087A1-20111208-C00130
Figure US20110301087A1-20111208-C00131
or a pharmaceutically acceptable salt of any of the foregoing.
11. The compound of claim 1, wherein P comprises at least three contiguous amino acid residues of the i1 loop.
12. The compound of claim 11, wherein P is derived from the following sequence: VLFLRLRSIRCLRNIIHWN (SEQ ID NO: 1).
13. The compound of claim 12, wherein P is a sequence selected from:
VLFLRLRSIRCLRNIIHWN; (SEQ ID NO: 1) VLFLRLRSIRCLRNIIHW; (SEQ ID NO: 2) GSGRLRSIRSLRNIIHWN; (SEQ ID NO: 3) VLFLRLRSIRCLRNIIH; (SEQ ID NO: 4) FLRLRSIRCLRNIIHWN; (SEQ ID NO: 5) GSGRLRSIRSLRNIIH; (SEQ ID NO: 6) LFIRIRSIRSLRNIIH; (SEQ ID NO: 7) VLFLRLRSIRCLRNII; (SEQ ID NO: 8) RLRSIRCLRNIIHWN; (SEQ ID NO: 9) RLRSIRSLRNIIHWN; (SEQ ID NO: 10) VLFLRLRSIRCLRNI; (SEQ ID NO: 11) VLFLRLRSIRCLRN; (SEQ ID NO: 12) RLRSIRSLRNIIH; (SEQ ID NO: 13) RSIRSLRNIIHWN; (SEQ ID NO: 14) VLFLRLRSIRCLR; (SEQ ID NO: 15) RSIRCLRNIIHW; (SEQ ID NO: 16) RSIRSLRNIIH; (SEQ ID NO: 17) RSIRCLRNIIH; (SEQ ID NO: 18) RLRSIRSLRN; (SEQ ID NO: 19) RSLRNIIHWN; (SEQ ID NO: 20) RLRSIRSLR; (SEQ ID NO: 21) RSLRNIIH; (SEQ ID NO: 22) and RSIRSLRN. (SEQ ID NO: 23)
14. The compound of claim 1, wherein is P derived from the i2 loop and is represented by the following structural formula or a pharmaceutically acceptable salt thereof
Y1-Y2-Y3-Y4-Y5-Y6-Y7-Y8-Y9-Y10-Y11-Y12-Y13-Y14-Y15-Y16- Y17-Y18-Y19-R1,
wherein:
Y1 is absent or a histidine residue;
Y2 is absent or a threonine residue;
Y3 is absent or an alanine residue;
Y4 is absent or an isoleucine or a glycine residue;
Y5 is absent or a valine or serine residue;
Y6 is absent or a leucine or glycine residue;
Y7 is absent or a threonine or serine residue;
Y8 is absent or a tyrosine or glycine residue;
Y9 is absent or serine, threonine or glycine residue;
Y10 is absent or a threonine, serine, tyrosine, glycine or alanine residue;
Y11 is absent or an aspartic acid, alanine, serine or glycine residue;
Y12 is absent or an arginine or alanine residue;
Y13 is absent or a leucine or an alanine residue;
Y14 is absent or an arginine or alanine residue;
Y15 is absent or a lysine or an alanine residue;
Y16 is absent or a tryptophan, methionine or an alanine residue;
Y17 is absent or a methionine, norleucine, phenylalanine or an alanine residue;
Y18 is absent or a phenylalanine, isoleucine or an alanine residue;
Y19 is absent or an isoleucine or alanine residue, provided that at least five of Y1-Y19, are present; and
R1 is OR2 or N(R2)2;
each R2 is independently hydrogen or (C1-C10)alkyl; and
from 0 to 5 amino acid residues are present in the D configuration.
15. The compound of claim 14, wherein at least three of Y7, Y8, Y9, Y10, Y11, Y12, Y13, Y14, Y15, Y16, Y17, Y18 and Y19 are present.
16. The compound of claim 15, wherein:
Y9 is serine, threonine or glycine residue;
Y10 is a threonine, serine, tyrosine, glycine or alanine residue; and
Y11 is an aspartic acid, alanine, serine or glycine residue.
17. The compound of claim 15, wherein:
Y12 is an arginine or alanine residue;
Y13 is a leucine or an alanine residue; and
Y14 is an arginine or alanine residue.
18. The compound of claim 16, wherein:
Y9 is serine, threonine or glycine residue;
Y10 is a threonine, serine, tyrosine, glycine or alanine residue;
Y11 is an aspartic acid, alanine, serine or glycine residue; and
Y12 is an arginine or alanine residue.
19. The compound of claim 17, wherein:
Y11 is an aspartic acid, alanine, serine or glycine residue;
Y12 is an arginine or alanine residue;
Y13 is a leucine or an alanine residue; and
Y14 is an arginine or alanine residue.
20. The compound of claim 18, wherein:
Y9 is serine, threonine or glycine residue;
Y10 is a threonine, serine, tyrosine, glycine or alanine residue;
Y11 is an aspartic acid, alanine, serine or glycine residue;
Y12 is an arginine or alanine residue; and
Y13 is a leucine or an alanine residue.
21. The compound of claim 19, wherein:
Y10 is a threonine, serine, tyrosine, glycine or alanine residue;
Y11 is an aspartic acid, alanine, serine or glycine residue;
Y12 is an arginine or alanine residue;
Y13 is a leucine or an alanine residue; and
Y14 is an arginine or alanine residue.
22. The compound of claim 20, wherein:
Y9 is serine, threonine or glycine residue;
Y10 is a threonine, serine, tyrosine, glycine or alanine residue;
Y11 is an aspartic acid, alanine, serine or glycine residue;
Y12 is an arginine or alanine residue;
Y13 is a leucine or an alanine residue; and
Y14 is an arginine or alanine residue.
23. The compound of claim 21, wherein:
Y9 is serine residue;
Y10 is a threonine residue;
Y11 is an aspartic acid residue;
Y12 is an arginine residue;
Y13 is a leucine residue; and
Y14 is an arginine residue.
24. The compound of claim 15, wherein:
Y7 is a threonine or serine residue;
Y8 is a tyrosine or glycine residue;
Y9 is a serine, threonine or glycine residue; and
Y10 is a threonine, serine, tyrosine, glycine or alanine residue.
25. The compound of claim 24, wherein:
Y7 is a threonine residue;
Y8 is a tyrosine residue;
Y9 is a serine residue; and
Y10 is a threonine residue.
26. The compound of claim 25, wherein Y11 is an aspartic acid, alanine, serine or glycine residue.
27. The compound of claim 26, wherein Y11 is an aspartic acid residue.
28. The compound of claim 15, wherein:
Y12 is an arginine or alanine residue;
Y13 is or a leucine or an alanine residue;
Y14 is an arginine or alanine residue; and
Y15 is a lysine or an alanine residue.
29. The compound of claim 28, wherein one of Y12, Y13, Y14, and Y15 is an alanine residue, provided that the other three of Y12, Y13, Y14, and Y15 are not an alanine residue.
30. The compound of claim 28, wherein:
Y12 is an arginine residue;
Y13 is a leucine residue;
Y14 is an arginine residue; and
Y15 is a lysine residue.
31. The compound of claim 28, wherein Y16 is a tryptophan, methionine or an alanine residue.
32. The compound of claim 31, wherein Y16 is a tryptophan residue.
33. The compound of claim 31, wherein Y16 is a methionine residue.
34. The compound of claim 15, wherein:
Y15 is a lysine or an alanine residue;
Y16 is a tryptophan, methionine or an alanine residue;
Y17 is a methionine, norleucine, phenylalanine or an alanine residue; and
Y18 is a phenylalanine, isoleucine or an alanine residue.
35. The compound of claim 34, wherein one of Y15, Y16, Y17, and Y18 is an alanine residue, provided that the other three of Y15, Y16, Y17, and Y18 are not an alanine residue.
36. The compound of claim 34, wherein:
Y15 is a lysine residue;
Y16 is a tryptophan or methionine residue;
Y17 is a methionine, norleucine, or phenylalanine residue; and
Y18 is a phenylalanine or isoleucine residue.
37. The compound of claim 36, wherein:
Y15 is a lysine residue;
Y16 is a tryptophan residue;
Y17 is a methionine residue; and
Y18 is a phenylalanine residue.
38. The compound of claim 34, wherein Y19 is an isoleucine or alanine residue.
39. The compound of claim 37, wherein Y19 is an isoleucine residue.
40. The compound of claim 14, selected from:
Figure US20110301087A1-20111208-C00132
Figure US20110301087A1-20111208-C00133
Figure US20110301087A1-20111208-C00134
Figure US20110301087A1-20111208-C00135
Figure US20110301087A1-20111208-C00136
Figure US20110301087A1-20111208-C00137
Figure US20110301087A1-20111208-C00138
Figure US20110301087A1-20111208-C00139
Figure US20110301087A1-20111208-C00140
Figure US20110301087A1-20111208-C00141
Figure US20110301087A1-20111208-C00142
Figure US20110301087A1-20111208-C00143
Figure US20110301087A1-20111208-C00144
Figure US20110301087A1-20111208-C00145
Figure US20110301087A1-20111208-C00146
Figure US20110301087A1-20111208-C00147
Figure US20110301087A1-20111208-C00148
Figure US20110301087A1-20111208-C00149
Figure US20110301087A1-20111208-C00150
Figure US20110301087A1-20111208-C00151
or a pharmaceutically acceptable salt of any of the foregoing.
41. The compound of claim 1, wherein P comprises at least three contiguous amino acid residues of the i2 loop.
42. The compound of claim 41, wherein P is derived from the following sequence:
HTAIVLTYSTDRLRKWMFI. (SEQ ID NO: 24)
43. The compound of claim 42, wherein P is a sequence selected from:
HTAIVLTYSTDRLRKWMFI; (SEQ ID NO: 24) HTAIVLTYSTDRLRKWM; (SEQ ID NO: 25) AIVLTYSTDRLRKWMFI; (SEQ ID NO: 26) GSGTYSTDRLRKWMFI; (SEQ ID NO: 27) HTAIVLTYSTDRLRK; (SEQ ID NO: 28) GSGTYSTDRLRKWMF; (SEQ ID NO: 29) VLTYSTDRLRKWMFI; (SEQ ID NO: 30) HTAIVLTYSTDRLR; (SEQ ID NO: 31) LTYSTDRLRKWMFI; (SEQ ID NO: 32) TYSTDRLRKWMFI; (SEQ ID NO: 33) HTAIVLTYSTDR; (SEQ ID NO: 34) GSGTYDRLRKWM; (SEQ ID NO: 35) GSGTYSTDRLRK; (SEQ ID NO: 36) GSTDRLRKWMFI; (SEQ ID NO: 37) GSTDRLRKWMFA; (SEQ ID NO: 38) GSTDRLRKWMAI; (SEQ ID NO: 39) GSTDRLRKWAFI; (SEQ ID NO: 40) GSTDRLRKAMFI; (SEQ ID NO: 41) GSTDRLRKAXFI; (SEQ ID NO: 42) YSTDRLRKWMFI; (SEQ ID NO: 43) GSTDRLRKMFI; (SEQ ID NO: 44) GSGRLRKWMFI; (SEQ ID NO: 45) STDRLRKWMFI; (SEQ ID NO: 46) YSTDRLRKWMF; (SEQ ID NO: 47) STDRLRKWMF; (SEQ ID NO: 48) ADRLRKWMFI; (SEQ ID NO: 49) GSRLRKWMFI; (SEQ ID NO: 50) TDRLRKWMFI; (SEQ ID NO: 51) TDRLRKWXFI; (SEQ ID NO: 52) TARLRKWMFI; (SEQ ID NO: 53) TDALRKWMFI; (SEQ ID NO: 54) TDRARKWMFI; (SEQ ID NO: 55) TDRLAKWMFI; (SEQ ID NO: 56) TDRLRAWMFI; (SEQ ID NO: 57) TDRLRKAMFI; (SEQ ID NO: 58) TDRLRKWAFI; (SEQ ID NO: 59) TDRLRKWMAI; (SEQ ID NO: 60) TDRLRKWMFA; (SEQ ID NO: 61) TYSTDRLRK; (SEQ ID NO: 62) STDRLRKMF; (SEQ ID NO: 63) GSGRLRKWM; (SEQ ID NO: 64) GRLRKWMFI; (SEQ ID NO: 65) DRLRKWMFI; (SEQ ID NO: 66) DRLRKWMAI; (SEQ ID NO: 67) GSTDRLRK; (SEQ ID NO: 68) RLRKWMFI; (SEQ ID NO: 69) and RLRKWMAI. (SEQ ID NO: 70)
44. The compound of claim 1, wherein P is derived from the i3 loop and is represented by the following structural formula or a pharmaceutically acceptable salt thereof:
Z1-Z2-Z3-Z4-Z5-Z6-Z7-Z8-Z9-Z10-Z11-Z12-Z13-Z14-Z15-Z16- Z17-Z18-Z19-Z20-Z21-Z22-Z23-Z24-Z25-Z26-Z27-Z28-Z29-Z30-R1,
wherein
Z1 is absent or a phenylalanine residue;
Z2 is absent or an asparagine residue;
Z3 is absent or an isoleucine residue;
Z4 is absent or a valine residue;
Z5 is absent or an arginine residue;
Z6 is absent or an isoleucine residue;
Z7 is absent or a leucine residue;
Z8 is absent or a methionine residue;
Z9 is absent or a threonine residue;
Z10 is absent or a lysine residue;
Z11 is absent or a leucine residue;
Z12 is absent or an arginine residue;
Z13 is absent or an alanine residue;
Z14 is absent or a serine residue;
Z15 is absent or a threonine residue;
Z16 is absent or a threonine residue;
Z17 is absent or a serine residue;
Z18 is absent or a glutamic acid residue;
Z19 is absent or a threonine residue;
Z20 is absent or an isoleucine residue;
Z21 is absent or a glutamine residue;
Z22 is absent or a tyrosine residue;
Z23 is absent or an arginine residue;
Z24 is absent or a lysine residue;
Z25 is absent or an alanine residue;
Z26 is absent or a valine residue;
Z27 is absent or a lysine residue;
Z28 is absent or an alanine or serine residue;
Z29 is absent or a threonine residue; and
Z30 is absent or a leucine residue;
provided that at least five of Z1-Z30 are present;
R1 is OR2 or N(R2)2;
each R2 is independently hydrogen or (C1-C10)alkyl; and
from 0 to 5 amino acid residues are present in the D configuration.
45. The compound of claim 44, wherein at least three of Z5, Z6, Z7, Z8, and Z9 are present.
46. The compound of claim 45, wherein:
Z5 is an arginine residue;
Z6 is an isoleucine residue; and
Z7 is a leucine residue.
47. The compound of claim 45, wherein:
Z7 is a leucine residue;
Z8 is a methionine residue; and
Z9 is a threonine residue.
48. The compound of claim 46, wherein:
Z5 is an arginine residue;
Z6 is an isoleucine residue;
Z7 is a leucine residue; and
Z8 is a methionine residue.
49. The compound of claim 47, wherein:
Z6 is an isoleucine residue;
Z7 is a leucine residue;
Z8 is absent or a methionine residue; and
Z9 is a threonine residue.
50. The compound of claim 48, wherein:
Z5 is an arginine residue;
Z6 is an isoleucine residue;
Z7 is a leucine residue;
Z8 is a methionine residue; and
Z9 is a threonine residue.
51. The compound of claim 44, wherein at least three of Z10, Z11, Z12, and Z13 are present.
52. The compound of claim 51, wherein:
Z10 is a lysine residue;
Z11 is a leucine residue; and
Z12 is an arginine residue.
53. The compound of claim 51, wherein:
Z11 is a leucine residue;
Z12 is an arginine residue; and
Z13 is an alanine residue.
54. The compound of claim 52, wherein:
Z10 is a lysine residue;
Z11 is a leucine residue;
Z12 is an arginine residue; and
Z13 is an alanine residue.
55. The compound of claim 44, wherein at least three of Z14, Z15, Z16, and Z17 are present.
56. The compound of claim 55, wherein:
Z14 is a serine residue;
Z15 is a threonine residue; and
Z16 is a threonine residue.
57. The compound of claim 55, wherein:
Z15 is a threonine residue;
Z16 is a threonine residue; and
Z17 is a serine residue.
58. The compound of claim 56, wherein:
Z14 is a serine residue;
Z15 is a threonine residue;
Z16 is a threonine residue; and
Z17 is a serine residue.
59. The compound of claim 44, wherein at least three of Z19, Z20, Z21, Z22, and Z23 are present.
60. The compound of claim 59, wherein:
Z19 is a threonine residue;
Z20 is an isoleucine residue; and
Z21 is a glutamine residue.
61. The compound of claim 59, wherein:
Z21 is a glutamine residue;
Z22 is a tyrosine residue; and
Z23 is an arginine residue.
62. The compound of claim 60, wherein:
Z19 is a threonine residue;
Z20 is an isoleucine residue;
Z21 is a glutamine residue; and
Z22 is a tyrosine residue.
63. The compound of claim 61, wherein:
Z20 is an isoleucine residue;
Z21 is a glutamine residue;
Z22 is a tyrosine residue; and
Z23 is an arginine residue.
64. The compound of claim 62, wherein:
Z19 is a threonine residue;
Z20 is an isoleucine residue;
Z21 is a glutamine residue;
Z22 is a tyrosine residue; and
Z23 is an arginine residue.
65. The compound of claim 44, wherein at least three of Z24, Z25, Z26, Z27, and Z28 are present.
66. The compound of claim 65, wherein:
Z24 is a lysine residue;
Z25 is an alanine residue; and
Z26 is a valine residue.
67. The compound of claim 65, wherein:
Z26 is a valine residue;
Z27 is a lysine residue; and
Z28 is an alanine or serine residue.
68. The compound of claim 66, wherein:
Z24 is a lysine residue;
Z25 is an alanine residue;
Z26 is a valine residue; and
Z27 is a lysine residue.
69. The compound of claim 67, wherein
Z25 is an alanine residue;
Z26 is a valine residue;
Z27 is a lysine residue; and
Z28 is an alanine or serine residue.
70. The compound of claim 68, wherein:
Z24 is a lysine residue;
Z25 is an alanine residue;
Z26 is a valine residue;
Z27 is a lysine residue; and
Z28 is an alanine or serine residue.
71. The compound of claim 70, wherein Z28 is an alanine residue.
72. The compound of claim 44, selected from:
Figure US20110301087A1-20111208-C00152
Figure US20110301087A1-20111208-C00153
Figure US20110301087A1-20111208-C00154
Figure US20110301087A1-20111208-C00155
or a pharmaceutically acceptable salt of any of the foregoing.
73. The compound of claim 1, wherein P comprises at least three contiguous amino acid residues of the i3 loop.
74. The compound of claim 73, wherein P is derived from the following sequence:
FNIVRILMTKLRASTTSETIQYRKAVKA. (SEQ ID NO: 71)
75. The compound of claim 74, wherein P is a sequence selected from:
FNIVRILMTKLRASTTSETIQYRKAVKA; (SEQ ID NO: 71) IVRILMTKLRASTTSETIQYRKAVKA; (SEQ ID NO: 72) FNIVRILMTKLRASTTSETIQYRKAV; (SEQ ID NO: 73) RILMTKLRASTTSETIQYRKAVKA; (SEQ ID NO: 74) FNIVRILMTKLRASTTSETIQYRK; (SEQ ID NO: 75) FNIVRILMTKLRASTTSETIQYR; (SEQ ID NO: 76) LMTKLRASTTSETIQYRKAVKA; (SEQ ID NO: 77) FNIVRILMTKLRASTTSETIQY; (SEQ ID NO: 78) TKLRASTTSETIQYRKAVKA; (SEQ ID NO: 79) LMTKLRASTTSETIQYRKAV; (SEQ ID NO: 80) LRASTTSETIQYRKAVKA; (SEQ ID NO: 81) LMTKLRASTTSETIQYRK; (SEQ ID NO: 82) LMTKLRASTTSETIQYR; (SEQ ID NO: 83) ASTTSETIQYRKAVKA; (SEQ ID NO: 84) ASTTSETIQYRKAVKS; (SEQ ID NO: 85) LMTKLRASTTSETIQ; (SEQ ID NO: 86) SETIQYRKAVKATL; (SEQ ID NO: 87) LMTKLRASTTSETI; (SEQ ID NO: 88) RILMTKLRASTTS; (SEQ ID NO: 89) SETIQYRKAVKA; (SEQ ID NO: 90) TKLRASTTSETI; (SEQ ID NO: 91) ASTTSETIQYR; (SEQ ID NO: 92) TKLRASTTSET; (SEQ ID NO: 93) KLRASTTSETI; (SEQ ID NO: 94) TIQYRKAVKA; (SEQ ID NO: 95) KLRASTTSET; (SEQ ID NO: 96) RILMTKLRA; (SEQ ID NO: 97) and KLRASTTSE. (SEQ ID NO: 98)
76. The compound of claim 1, wherein P derived from the i4 domain and is represented by the following structural formula or a pharmaceutically acceptable salt thereof
M1-M2-M3-M4-M5-M6-M7-M8-M9-M10-M11-M12-M13-M14-M15-M16- M17-M18-M19-M20-M21-M22-M23-M24-M25-M26-M27-M28-M29-M30- M31-M32-M33-M34-M35-M36-M37-M38-M39-M40-M41-M42-M43-M44- M45-M46-M47-R1,
wherein:
M1 is absent or a glutamic acid residue;
M2 is absent or a valine residue;
M3 is absent or an arginine residue;
M4 is absent or a serine residue;
M5 is absent or an alanine residue;
M6 is absent or an isoleucine residue;
M7 is absent or an arginine residue;
M8 is absent or a lysine residue;
M9 is absent or an arginine residue;
M10 is absent or a tryptophan residue;
M11 is absent or a histidine residue;
M12 is absent or an arginine residue;
M13 is absent or a tryptophan residue;
M14 is absent or a glutamine residue;
M15 is absent or an aspartic acid residue;
M16 is absent or a lysine residue;
M17 is absent or a histidine or glycine residue;
M18 is absent or a serine residue;
M19 is absent or an isoleucine residue;
M20 is absent or an arginine residue;
M21 is absent or an alanine residue;
M22 is absent or an arginine residue;
M23 is absent or a valine residue;
M24 is absent or an alanine residue;
M25 is absent or an arginine residue;
M26 is absent or an alanine residue;
M27 is absent or a methionine residue;
M28 is absent or a serine residue;
M29 is absent or an isoleucine residue;
M30 is absent or a proline residue;
M31 is absent or a threonine residue;
M32 is absent or a serine residue;
M33 is absent or a proline residue;
M34 is absent or a threonine residue;
M35 is absent or an arginine residue;
M36 is absent or a valine residue;
M37 is absent or a serine residue;
M38 is absent or a phenylalanine residue;
M39 is absent or a histidine residue;
M40 is absent or a serine residue;
M41 is absent or an isoleucine residue;
M42 is absent or a lysine residue;
M43 is absent or a glutamine residue;
M44 is absent or a serine residue;
M45 is absent or a threonine residue;
M46 is absent or an alanine residue; and
M47 is absent or a valine residue; provided that at least five of M1-M47 are present;
R1 is OR2 or N(R2)2;
each R2 is independently hydrogen or (C1-C10)alkyl; and
from 0 to 5 amino acid residues are present in the D configuration.
77. The compound of claim 76, wherein at least three of M2-M7 are present.
78. The compound of claim 77, wherein:
M2 is a valine residue;
M3 is an arginine residue; and
M4 is a serine residue.
79. The compound of claim 77, wherein:
M5 is an alanine residue;
M6 is an isoleucine residue; and
M7 is an arginine residue.
80. The compound of claim 78, wherein:
M2 is a valine residue;
M3 is an arginine residue;
M4 is a serine residue; and
M5 is an alanine residue.
81. The compound of claim 79, wherein:
M4 is a serine residue;
M5 is an alanine residue;
M6 is an isoleucine residue; and
M7 is an arginine residue.
82. The compound of claim 80, wherein:
M2 is a valine residue;
M3 is an arginine residue;
M4 is a serine residue;
M5 is an alanine residue;
M6 is an isoleucine residue.
83. The compound of claim 81, wherein:
M3 is an arginine residue;
M4 is a serine residue;
M5 is an alanine residue;
M6 is an isoleucine residue; and
M7 is an arginine residue.
84. The compound of claim 82, wherein:
M2 is a valine residue;
M3 is an arginine residue;
M4 is a serine residue;
M5 is an alanine residue;
M6 is an isoleucine residue; and
M7 is an arginine residue.
85. The compound of claim 76, wherein at least three of M9-M13 are present.
86. The compound of claim 85, wherein:
M9 is an arginine residue;
M10 is a tryptophan residue;
M11 is a histidine residue;
M12 is an arginine residue.
87. The compound of claim 85, wherein:
M10 is a tryptophan residue;
M11 is a histidine residue;
M12 is an arginine residue; and
M13 is a tryptophan residue.
88. The compound of claim 86, wherein:
M9 is an arginine residue;
M10 is a tryptophan residue;
M11 is a histidine residue;
M12 is an arginine residue; and
M13 is a tryptophan residue.
89. The compound of claim 76, wherein at least three of M18-M22 are present.
90. The compound of claim 89, wherein:
M18 is a serine residue;
M19 is an isoleucine residue;
M20 is an arginine residue; and
M21 is an alanine residue.
91. The compound of claim 89, wherein:
M19 is an isoleucine residue;
M20 is an arginine residue;
M21 is an alanine residue; and
M22 is an arginine residue.
92. The compound of claim 90, wherein:
M18 is a serine residue;
M19 is an isoleucine residue;
M20 is an arginine residue;
M21 is an alanine residue; and
M22 is an arginine residue.
93. The compound of claim 76, wherein at least three of M23-M27 are present.
94. The compound of claim 93, wherein:
M23 is a valine residue;
M24 is an alanine residue;
M25 is an arginine residue; and
M26 is an alanine residue.
95. The compound of claim 93, wherein:
M24 is an alanine residue;
M25 is an arginine residue;
M26 is an alanine residue;
M27 is a methionine residue.
96. The compound of claim 94, wherein:
M23 is a valine residue;
M24 is an alanine residue;
M25 is an arginine residue;
M26 is an alanine residue;
M27 is a methionine residue.
97. The compound of claim 76, wherein at least three of M25-M29 are present.
98. The compound of claim 97, wherein:
M25 is an arginine residue;
M26 is an alanine residue;
M27 is a methionine residue; and
M28 is a serine residue.
99. The compound of claim 97, wherein:
M26 is an alanine residue;
M27 is a methionine residue;
M28 is a serine residue; and
M29 is an isoleucine residue.
100. The compound of claim 98, wherein:
M25 is an arginine residue;
M26 is an alanine residue;
M27 is a methionine residue;
M28 is a serine residue; and
M29 is an isoleucine residue.
101. The compound of claim 76, wherein at least three of M32-M36 are present.
102. The compound of claim 101, wherein:
M32 is a serine residue;
M33 is a proline residue;
M34 is a threonine residue; and
M35 is an arginine residue.
103. The compound of claim 101, wherein:
M33 is a proline residue;
M34 is a threonine residue;
M35 is an arginine residue; and
M36 is a valine residue.
104. The compound of claim 102, wherein:
M32 is a serine residue;
M33 is a proline residue;
M34 is a threonine residue;
M35 is an arginine residue; and
M36 is a valine residue.
105. The compound of claim 76, selected from:
Figure US20110301087A1-20111208-C00156
or a pharmaceutically acceptable salt of any of the foregoing.
106. The compound of claim 1, wherein P comprises at least three contiguous amino acid residues of the i4 domain.
107. The compound of claim 106, wherein P is derived from the following sequence: EVRSAIRKRWHRWQDKHSIRARVARAMSIPTSPTRVSFHSIKQSTAV (SEQ ID NO: 99).
108. The compound of claim 107, wherein P is a sequence selected from
RAMSIPTSPTRVSFHSIKQSTAV; (SEQ ID NO: 100) HSIRARVARAMSIPTSPTRV; (SEQ ID NO: 101) RWQDKHSIRARVARAMSI; (SEQ ID NO: 102) EVRSAIRKRWHRWQD; (SEQ ID NO: 103) RWHRWQDKHSIRAR; (SEQ ID NO: 104) VRSAIRKRWHRW; (SEQ ID NO: 105) and GSIRARVARAM. (SEQ ID NO: 106)
109. The compound of claim 1, wherein T is an optionally substituted (C6-C30)alkyl, (C6-C30)alkenyl, (C6-C30)alkynyl, wherein 0-3 carbon atoms are replaced with oxygen, sulfur, nitrogen or a combination thereof.
110. The compound of claim 109, wherein T is selected from the group consisting of: CH3(CH2)16, CH3(CH2)15, CH3(CH2)14, CH3(CH2)13, CH3(CH2)12, CH3(CH2)11, CH3(CH2)10, CH3(CH2)9, CH3(CH2)8, CH3(CH2)9OPh-, CH3(CH2)6C═C(CH2)6, CH3(CH2)11O(CH2)3, and CH3(CH2)9O(CH2)2.
111. The compound of claim 1, wherein T is a fatty acid derivative.
112. The compound of claim 111, wherein the fatty acid is selected from the group consisting of: butyric acid, caproic acid, caprylic acid, capric acid, lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, lignoceric acid, myristoleic acid, palmitoleic acid, oleic acid, linoleic acid, α-linolenic acid, arachidonic acid, eicosapentaenoic acid, erucic acid, docosahexaenoic acid.
113. The compound of claim 1, wherein T is a bile acid derivative.
114. The compound of claim 113, wherein the bile acid is selected from the group consisting of: lithocholic acid, chenodeoxycholic acid, deoxycholic acid, cholanic acid, cholic acid, ursocholic acid, ursodeoxycholic acid, isoursodeoxycholic acid, lagodeoxycholic acid, dehydrocholic acid, hyocholic acid, and hyodeoxycholic acid.
115. The compound of claim 1, wherein T is selected from sterols; progestagens; glucocorticoids; mineralcorticoids; androgens; and estrogens.
116. The compound of claim 1, wherein TL is selected from:
CH3(CH2)15—C(O);
CH3(CH2)13—C(O);
CH3(CH2)9O(CH2)2C(O);
CH3(CH2)10O(CH2)2C(O);
CH3(CH2)6C═C(CH2)6—C(O);
LCA-C(O); and
CH3(CH2)9OPh-C(O) wherein
Figure US20110301087A1-20111208-C00157
117. A method of treating diseases and conditions associated with CRF1 modulation in a patient in need thereof comprising administering to said patient and effective amount of a compound of claim 1.
118. The method of claim 117, wherein the disease or condition is selected from: peripherally acting inflammatory bowel disease, irritable bowel syndrome, stress response, anxiety, sleep disorder, addictive behavior, acute and chronic neurodegeneration, preterm labor and pain.
119. A pharmaceutical composition comprising a compound of claim 1, and a pharmaceutically acceptable carrier.
US13/127,602 2008-11-04 2009-11-04 Crf1 receptor compounds Abandoned US20110301087A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/127,602 US20110301087A1 (en) 2008-11-04 2009-11-04 Crf1 receptor compounds

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US19819408P 2008-11-04 2008-11-04
PCT/US2009/005975 WO2010053546A2 (en) 2008-11-04 2009-11-04 Crf1 receptor compounds
US13/127,602 US20110301087A1 (en) 2008-11-04 2009-11-04 Crf1 receptor compounds

Publications (1)

Publication Number Publication Date
US20110301087A1 true US20110301087A1 (en) 2011-12-08

Family

ID=42153453

Family Applications (1)

Application Number Title Priority Date Filing Date
US13/127,602 Abandoned US20110301087A1 (en) 2008-11-04 2009-11-04 Crf1 receptor compounds

Country Status (2)

Country Link
US (1) US20110301087A1 (en)
WO (1) WO2010053546A2 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8933059B2 (en) 2012-06-18 2015-01-13 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US8987237B2 (en) 2011-11-23 2015-03-24 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9180091B2 (en) 2012-12-21 2015-11-10 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US9289382B2 (en) 2012-06-18 2016-03-22 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US9931349B2 (en) 2016-04-01 2018-04-03 Therapeuticsmd, Inc. Steroid hormone pharmaceutical composition
US10052386B2 (en) 2012-06-18 2018-08-21 Therapeuticsmd, Inc. Progesterone formulations
US10206932B2 (en) 2014-05-22 2019-02-19 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10258630B2 (en) 2014-10-22 2019-04-16 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10286077B2 (en) 2016-04-01 2019-05-14 Therapeuticsmd, Inc. Steroid hormone compositions in medium chain oils
US10328087B2 (en) 2015-07-23 2019-06-25 Therapeuticsmd, Inc. Formulations for solubilizing hormones
US10471148B2 (en) 2012-06-18 2019-11-12 Therapeuticsmd, Inc. Progesterone formulations having a desirable PK profile
US10471072B2 (en) 2012-12-21 2019-11-12 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10537581B2 (en) 2012-12-21 2020-01-21 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10806740B2 (en) 2012-06-18 2020-10-20 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11077108B2 (en) 2016-09-07 2021-08-03 The Regents Of The University Of California Allosteric corticotropin-releasing factor receptor 1 (CRFR1) antagonists that decrease p-tau and improve cognition
US11246875B2 (en) 2012-12-21 2022-02-15 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11266661B2 (en) 2012-12-21 2022-03-08 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2001257169C1 (en) * 2000-04-21 2011-06-16 Tufts Medical Center, Inc. G-protein coupled receptor (GPCR) agonists and antagonists and methods of activating and inhibiting GPCR using the same
WO2003024990A2 (en) * 2001-09-14 2003-03-27 The University Of Tennessee Research Corporation Variants of corticotropin releasing hormone receptor type 1 and uses thereof
WO2006052723A2 (en) * 2004-11-04 2006-05-18 New England Medical Center Hospitals, Inc. G protein coupled receptor agonists and antagonists and methods of use
EP2114437A2 (en) * 2006-10-16 2009-11-11 ConjuChem Biotechnologies Inc. Modified corticotropin releasing factor peptides and uses thereof

Cited By (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9248136B2 (en) 2011-11-23 2016-02-02 Therapeuticsmd, Inc. Transdermal hormone replacement therapies
US8987237B2 (en) 2011-11-23 2015-03-24 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11793819B2 (en) 2011-11-23 2023-10-24 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US8993549B2 (en) 2011-11-23 2015-03-31 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US8993548B2 (en) 2011-11-23 2015-03-31 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11103516B2 (en) 2011-11-23 2021-08-31 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10675288B2 (en) 2011-11-23 2020-06-09 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9114146B2 (en) 2011-11-23 2015-08-25 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9114145B2 (en) 2011-11-23 2015-08-25 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10639375B2 (en) 2012-06-18 2020-05-05 Therapeuticsmd, Inc. Progesterone formulations
US8933059B2 (en) 2012-06-18 2015-01-13 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9289382B2 (en) 2012-06-18 2016-03-22 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US9301920B2 (en) 2012-06-18 2016-04-05 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11865179B2 (en) 2012-06-18 2024-01-09 Therapeuticsmd, Inc. Progesterone formulations having a desirable PK profile
US10052386B2 (en) 2012-06-18 2018-08-21 Therapeuticsmd, Inc. Progesterone formulations
US8987238B2 (en) 2012-06-18 2015-03-24 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11529360B2 (en) 2012-06-18 2022-12-20 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11166963B2 (en) 2012-06-18 2021-11-09 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11110099B2 (en) 2012-06-18 2021-09-07 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9006222B2 (en) 2012-06-18 2015-04-14 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10471148B2 (en) 2012-06-18 2019-11-12 Therapeuticsmd, Inc. Progesterone formulations having a desirable PK profile
US11033626B2 (en) 2012-06-18 2021-06-15 Therapeuticsmd, Inc. Progesterone formulations having a desirable pk profile
US10806740B2 (en) 2012-06-18 2020-10-20 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9012434B2 (en) 2012-06-18 2015-04-21 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10835487B2 (en) 2012-12-21 2020-11-17 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11246875B2 (en) 2012-12-21 2022-02-15 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11622933B2 (en) 2012-12-21 2023-04-11 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US10537581B2 (en) 2012-12-21 2020-01-21 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10806697B2 (en) 2012-12-21 2020-10-20 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11497709B2 (en) 2012-12-21 2022-11-15 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US9180091B2 (en) 2012-12-21 2015-11-10 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US10888516B2 (en) 2012-12-21 2021-01-12 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US11351182B2 (en) 2012-12-21 2022-06-07 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10471072B2 (en) 2012-12-21 2019-11-12 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11065197B2 (en) 2012-12-21 2021-07-20 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US11304959B2 (en) 2012-12-21 2022-04-19 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11266661B2 (en) 2012-12-21 2022-03-08 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10568891B2 (en) 2012-12-21 2020-02-25 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11241445B2 (en) 2012-12-21 2022-02-08 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11116717B2 (en) 2012-12-21 2021-09-14 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US11123283B2 (en) 2012-12-21 2021-09-21 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US11103513B2 (en) 2014-05-22 2021-08-31 TherapeuticsMD Natural combination hormone replacement formulations and therapies
US10206932B2 (en) 2014-05-22 2019-02-19 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10398708B2 (en) 2014-10-22 2019-09-03 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10258630B2 (en) 2014-10-22 2019-04-16 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10668082B2 (en) 2014-10-22 2020-06-02 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10328087B2 (en) 2015-07-23 2019-06-25 Therapeuticsmd, Inc. Formulations for solubilizing hormones
US10912783B2 (en) 2015-07-23 2021-02-09 Therapeuticsmd, Inc. Formulations for solubilizing hormones
US10286077B2 (en) 2016-04-01 2019-05-14 Therapeuticsmd, Inc. Steroid hormone compositions in medium chain oils
US10532059B2 (en) 2016-04-01 2020-01-14 Therapeuticsmd, Inc. Steroid hormone pharmaceutical composition
US9931349B2 (en) 2016-04-01 2018-04-03 Therapeuticsmd, Inc. Steroid hormone pharmaceutical composition
US11077108B2 (en) 2016-09-07 2021-08-03 The Regents Of The University Of California Allosteric corticotropin-releasing factor receptor 1 (CRFR1) antagonists that decrease p-tau and improve cognition

Also Published As

Publication number Publication date
WO2010053546A3 (en) 2010-07-15
WO2010053546A2 (en) 2010-05-14

Similar Documents

Publication Publication Date Title
US20110301087A1 (en) Crf1 receptor compounds
US20110294738A1 (en) Pthr1 receptor compounds
US9096646B2 (en) CXCR4 receptor compounds
US20110300167A1 (en) Cxcr5 receptor compounds
US20120028888A1 (en) Apj receptor compounds
US20150011466A1 (en) APJ Receptor Compounds
US20220048967A1 (en) Gip/glp1 co-agonist compounds
CN103596972A (en) Polypeptides
US9155795B2 (en) CXCR4 receptor compounds
WO2021126990A1 (en) Oxytocin derivatives with improved properties
US20160159861A1 (en) APJ Receptor Compounds
Jin et al. Structure-based design, synthesis, and activity of peptide inhibitors of RGS4 GAP activity
Harsha Vardhan Reddy Design, Synthesis and Biological testing of Novel ligands for Ghrelin Receptor

Legal Events

Date Code Title Description
AS Assignment

Owner name: ANCHOR THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MCBRIDE, EDWARD;KULIOPULOS, ATHAN;MCMURRY, THOMAS J.;SIGNING DATES FROM 20100920 TO 20101019;REEL/FRAME:025195/0923

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION